US6337200B1 - Human telomerase catalytic subunit variants - Google Patents

Human telomerase catalytic subunit variants Download PDF

Info

Publication number
US6337200B1
US6337200B1 US09/128,354 US12835498A US6337200B1 US 6337200 B1 US6337200 B1 US 6337200B1 US 12835498 A US12835498 A US 12835498A US 6337200 B1 US6337200 B1 US 6337200B1
Authority
US
United States
Prior art keywords
leu
arg
ala
val
ser
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US09/128,354
Inventor
Gregg B. Morin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Geron Corp
University of Colorado
Original Assignee
Geron Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US09/128,354 priority Critical patent/US6337200B1/en
Application filed by Geron Corp filed Critical Geron Corp
Assigned to GERON CORPORATION reassignment GERON CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MORIN, GREGG B.
Priority to PCT/US1999/007097 priority patent/WO1999050386A2/en
Priority to AU33752/99A priority patent/AU3375299A/en
Priority to US09/990,080 priority patent/US7091021B2/en
Publication of US6337200B1 publication Critical patent/US6337200B1/en
Application granted granted Critical
Assigned to THE REGENTS OF THE UNIVERSITY OF COLORADO, GERON CORPORATION reassignment THE REGENTS OF THE UNIVERSITY OF COLORADO ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GERON CORPORATION
Priority to US11/504,402 priority patent/US8796438B2/en
Priority to US14/317,418 priority patent/US20150087064A1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense

Definitions

  • the present invention is related to the catalytic protein subunit of human telomerase.
  • the invention provides methods and compositions relating to medicine, molecular biology, chemistry, pharmacology, and medical diagnostic and prognostic technology.
  • telomeres the protein-DNA structures physically located on the ends of chromosomes, is thought to account for the phenomenon of cellular senescence or aging of normal human somatic cells in vitro and in vivo.
  • the maintenance of telomeres is a function of a telomere-specific DNA polymerase known as telomerase.
  • Telomerase is a ribonucleoprotein (RNP) that uses a portion of its RNA moiety as a template for telomeric DNA synthesis (Morin, 1997, Eur. J. Cancer 33:750).
  • telomere length and integrity of telomeres and the telomerase expression status of a cell is thus related to entry of a cell into a senescent stage (i.e., loss of proliferative capacity), or the ability of a cell to escape senescence, i.e., to become immortal.
  • telomere activity is detected in immortal cell lines and an extraordinarily diverse set of tumor tissues, but is not detected (i.e., was absent or below the assay threshold) in normal somatic cell cultures or normal tissues adjacent to a tumor (see, U.S. Pat. Nos. 5,629,154; 5,489,508; 5,648,215; and 5,639,613; see also, Morin, 1989, Cell 59: 521; Shay and Bacchetti 1997, Eur. J.
  • telomere activity in a tumor has been reported (e.g., U.S. Pat. No. 5,639,613, supra; Langford et al., 1997, Hum. Pathol. 28:416).
  • human telomerase is an ideal target for diagnosing and treating human diseases relating to cellular proliferation and senescence, such as cancer, or for increasing the proliferative capacity of a cell.
  • the invention provides an isolated or recombinant hTRT polypeptide that has telomerase catalytic activity.
  • the hTRT polypeptide has a deletion of at least 25 residues in the regions corresponding to residues 192-323, 200-323, 192-271, 200-271, 222-240, 415-450, 192-323 and 415-450, or 192-271 and 415-450 of hTRT.
  • residues 192-323, 200-323, 192-271, 200-271, 222-240, 415-450, 192-323 and 415-450, or 192-271 and 415-450 of hTRT are deleted.
  • the invention also provides a polynucleotide comprising a nucleotide sequence encoding these hTRT polypeptides.
  • the polynucleotide includes a promoter sequence operably linked to the nucleotide sequence encoding the hTRT polypeptide.
  • the invention also provides a method of preparing recombinant telomerase by contacting a recombinant hTRT polypeptide containing a deletion as described supra with a telomerase RNA component under conditions such that the recombinant protein and the telomerase RNA component associate to form a telomerase enzyme capable of catalyzing the addition of nucleotides to a telomerase substrate.
  • the hTRT polypeptide may be produced in an in vitro expression system and/or may be purified before the contacting step. In some embodiments, the contacting occurs in a cell.
  • the invention further provides a method for increasing the proliferative capacity of a vertebrate cell by introducing into a cell the recombinant hTRT polynucleotide encoding an hTRT deletion variant described supra.
  • the invention provides a cell, such as a human cell or other mammalian cell, comprising a nucleotide sequence that encodes the hTRT deletion variant polypeptide.
  • the invention provides such cells that have an increased proliferative capacity relative to a cell that is otherwise identical but does not comprise the recombinant polynucleotide.
  • an isolated or recombinant hTRT polypeptide that has a deletion of amino acid residues 192-450, 560-565, 637-660, 638-660, 748-766, 748-764, or 1055-1071, where the residue numbering is with reference to the hTRT polypeptide having the sequence provided in FIG. 1, is provided.
  • the invention provides an isolated, recombinant, or substantially purified polynucleotide encoding this polypeptide, which in some embodiments includes a promoter sequence operably linked to the nucleotide sequence encoding the hTRT polypeptide.
  • the invention also provides a method of reducing telomerase activity in a cell by introducing the polynucleotide described supra (i.e., having a deletion of deletion of amino acid residues 192-450, 560-565, 637-660, 638-660, 748-766, 748-764, or 1055-1071) into a cell under conditions in which it is expressed.
  • the hTRT polypeptide has one or more mutations other than, or in addition to, a deletion of at least 25 residues in the regions corresponding to residues 192-323, 200-323, 192-271, 200-271, 222-240, 415-450, 192-323 and 415-450, or 192-271 and 415-450 of hTRT.
  • FIG. 1 shows the amino acid sequence of a 1132-residue human telomerase reverse transcriptase (hTRT) protein (SEQ ID NO:2).
  • FIG. 2 shows the nucleotide sequence of a naturally occurring cDNA encoding the hTRT protein (SEQ ID NO:1).
  • Telomerase is a ribonucleoprotein complex (RNP) comprising an RNA component and a catalytic protein component.
  • the catalytic protein component of human telomerase hereinafter referred to as telomerase reverse transcriptase (“hTRT”), has been cloned, and protein, cDNA and genomic sequences determined. See, e.g., Nakamura et al., 1997, Science 277:955, and copending U.S. patent application Ser. No. 08/912,951 filed Aug. 14, 1997, still pending and Ser. No. 08/974,549, filed Nov. 19, 1997, now U.S. Pat. No. 6,166,178.
  • hTRT The sequence of a full-length native hTRT has been deposited in GenBank (Accession No. AF015950), and plasmid and phage vectors having hTRT coding sequences have been deposited with the American Type Culture Collection, Rockville, Md. (accession numbers 209024, 209016, and 98505).
  • the catalytic subunit protein of human telomerase has also been referred to as “hEST2” (Meyerson et al., 1997, Cell 90:785), “hTCS1” (Kilian et al., 1997, Hum. Mol. Genet. 6:2011), “TP2” (Harrington et al., 1997, Genes Dev.
  • hTERT Human TRT is also described in the aforereferenced priority applications and U.S. patent application Ser. No. 08/846,017, now abandoned, Ser. No. 08/844,419, now abandoned, and Ser. No. 08/724,643,now abandoned.
  • the RNA component of human telomerase (hTR) has also been characterized (see U.S. Pat. No. 5,583,016). All of the aforementioned applications and publications are incorporated by reference herein in their entirety and for all purposes.
  • telomere activity in human cells and other mammalian cells correlates with cell proliferative capacity, cell immortality, and the development of a neoplastic phenotype.
  • hTRT polypeptides including the hTRT variants described herein, and polynucleotides encoding hTRT polypeptides, are used, inter alia for conferring a telomerase activity (e.g., telomerase catalytic activity, infra) in a telomerase-negative cell such as a cell from a human, a mammal, a vertebrate, or other eukaryote ( see, e.g., Bodnar et al., 1998, Science 279:349 and copending U.S.
  • a telomerase activity e.g., telomerase catalytic activity, infra
  • a telomerase-negative cell such as a cell from a human, a mammal,
  • variants that lack at least one hTRT activity are used, inter alia, to inhibit telomerase activity in a cell (e.g., by acting as “dominant negative mutants”).
  • hTRT variants and polynucleotides encoding them, as described herein, are similarly useful in screening assays for identifying agents that modulate telomerase activity.
  • the hTRT variants of the present invention are characterized by one or more deletions or mutations, relative to a naturally occurring hTRT polypeptide, in defined regions of the protein, as described in detail infra. These hTRT variants may have none, one, or several of the biological activities that may be found in naturally-occurring full-length hTRT proteins.
  • telomerase catalytic activity the ability to extend a DNA primer that functions as a telomerase substrate by adding a partial, one, or more than one repeat of a sequence, e.g., TTAGGG, encoded by a template nucleic acid, e.g., hTR
  • telomerase conventional reverse transcriptase activity see Morin, 1997, supra, and Spence et al., 1995, Science 267:988
  • nucleolytic activity see Morin, 1997, supra; Collins and Grieder, 1993, Genes and Development 7:1364; Joyce and Steitz, 1987, Trends Biochem. Sci.
  • telomere binding activity see, Morin, 1997, supra; Collins et al., 1995, Cell 81:677; Harrington et al., 1995, J. Biol. Chem. 270:8893; dNTP binding activity (Morin, 1997, supra; Spence et al., supra); and RNA (e.g.. hTRT) binding activity (see Morin, 1997, supra; Harrington et al., 1997, Science 275:973; Collins et al., 1995, Cell 81:677).
  • the hTRT variant has telomerase catalytic activity.
  • Telomerase catalytic activity may be processive or nonprocessive.
  • Processive telomerase catalytic activity occurs when a telomerase RNP adds multiple repeats to a primer or telomerase before the DNA is released by the enzyme complex (see, e.g., Morin, 1989, Cell 59:521 and Morin, 1997, Eur. J. Cancer 33:750).
  • Nonprocessive activity occurs when telomerase adds a partial, or only one, repeat to a primer and is then released (see Morin, 1997, supra).
  • the hTRT variant has processive telomerase catalytic activity.
  • telomerase catalytic activity can be assayed by a variety of methods, including the “conventional assay” (Morin, 1989, Cell 59:521), the TRAP assay (U.S. Pat. No. 5,629,154; see also, PCT publication WO 97/15687, PCT publication WO 95/13381; Krupp et al. Nucleic Acids Res., 1997, 25: 919; Wright et al., 1995, Nuc. Acids Res. 23:3794), the “dot blot immunoassay” (U.S. patent application Ser. No. 08/833,377 now U.S. Pat. No.
  • telomere substrate used in these assays may have a natural telomere sequence, or may be have a synthetic oligonucleotide with a different sequence (see, e.g., Morin, 1989, Cell 59:521; Morin, 1991, Nature 353:454-56).
  • an hTRT variant is considered to have a specified activity if the activity is exhibited by either the hTRT variant polypeptide without an associated hTR RNA or in an hTRT-hTR complex.
  • Each of the hTRT activities described supra is also described in detail in copending U.S. patent applications Ser. No. 08/912,951 still pending and 08/974,549, now U.S. Pat. No. 6,166,178.
  • hTRT protein can be mutated (e.g., deleted) without loss of telomerase catalytic activity.
  • Sites of mutation e.g., deletion
  • hTRT polypeptides e.g., naturally occurring variants such as polymorphic variants, hTRT fusion proteins, hTRT homologs (e.g., from non-human species), and the like.
  • residues of the full-length hTRT protein having a sequence as provided in FIG. 1 are referred to herein by number, with the amino-terminal methionine (M) in FIG. 1 numbered “1”, and the carboxy-terminal aspartic acid (D) numbered “1132”.
  • Regions of the hTRT protein that can be mutated (e.g., deleted) without abolishing telomerase catalytic activity include the regions from amino acid residues 192 to 323 (inclusive) and residues 415 to 450 (inclusive). As is demonstrated in the experiments described infra, all or part of either of these regions, or all or part of both of them, can be deleted without abolishing the telomerase catalytic activity of the protein.
  • the regions from amino acid residues 192 to 323 and residues 415 to 450 may be referred to as “nonessential” regions of hTRT (i.e., not essential for telomerase catalytic activity).
  • the hTRT variants of the invention comprise deletions of, or other mutations in, these nonessential regions of hTRT.
  • certain mutations e.g., deletion of residues 415-450
  • the mutation is a deletion of at least one, typically at least about 10, and often at least about 25, at least about 50, or at least about 100 amino acid residues relative to a naturally occurring hTRT.
  • the mutation is a single amino acid substitution in a “non-essential” region, or a combinations of substitutions. Substitutions may be conservative substitutions or non-conservative substitutions.
  • the mutation is an insertion or substitution of amino acids, for example the insertion of residues that encode an epitope tag or novel proteolytic site.
  • Substitutions may be of one or more (e.g., all) of the residues in the above-mentioned regions or may be combined with deletions so that, e.g., a shorter heterologous sequence is a substituted for a longer hTRT sequence. It will be appreciated, as noted supra, that in some embodiments the hTRT variant has more than one different type of mutation relative to a naturally occurring hTRT protein (e.g., a deletion and a point mutation).
  • the hTRT variants of the invention have certain advantages compared to naturally occurring hTRT proteins.
  • mutations may confer more efficient in vitro expression of active hTRT (e.g., in expression systems in which shorter polypeptides are more efficiently expressed than longer polypeptides), may provide sequences that aid in purification (e.g., an epitope tag sequence), or may add a new functional moiety to the hTRT polypeptide (e.g., a 3′ ⁇ 5′ exonuclease domain from DNA polymerase I).
  • hTRT variant polypeptides of the invention comprising mutations (e.g., deletions) in the “non-essential” regions of the hTRT retain telomerase catalytic activity.
  • mutations e.g., deletions
  • polynucleotides that encode them are useful in any application for which other catalytically active hTRT proteins (e.g., wild-type hTRT proteins) or polynucleotides may be used, including, inter alia, in therapeutic, diagnostic, and screening uses.
  • Exemplary uses of hTRT polypeptides and polynucleotides are described in additional detail in the afore cited copending applications (e.g., U.S. Ser. No. 08/912,951, still pending and Ser. No. 08/974,549,now U.S. Pat. No. 6,166,178).
  • the hTRT variant of the invention is used to increase the proliferative capacity of a cell by, e.g., increasing telomerase activity in the cell (see, Bodnar et al. supra, and copending U.S. patent application Ser. No. 08/912,951, still pending and Ser. No. 08/974,549, now U.S. Pat. No. 6,166,178 for a detailed description of exemplary methods).
  • a polynucleotide comprising (i) a sequence encoding the hTRT variant polypeptide; (ii) an operably linked promoter (e.g., a heterologous promoter); and, (iii) optionally polyadenylation and termination signals, enhancers, or other regulatory elements, is introduced into a target cell (e.g., by transfection, lipofection, electroporation, or any other suitable method) under conditions in which the hTRT variant polypeptide is expressed.
  • the expression in the cell of the catalytically active hTRT variant of the invention results in increased proliferative capacity (e.g., an immortal phenotype).
  • the hTRT variant is used for in vitro reconstitution (IVR) of a telomerase ribonucleoprotein (e.g., comprising the hTRT variant polypeptide and a template RNA, e.g., hTR) that has telomerase catalytic activity.
  • IVR in vitro reconstitution
  • a telomerase ribonucleoprotein e.g., comprising the hTRT variant polypeptide and a template RNA, e.g., hTR
  • IVR in vitro reconstitution
  • an expression vector encoding an hTRT variant of the invention is expressed in an in vitro expression system (e.g., a coupled transcription-translation reticulocyte lysate system such as that described in U.S. Pat. No. 5,324,637).
  • the hTRT variant polypeptide is coexpressed with hTR.
  • the hTRT variant and hTR are separately expressed and then combined (mixed) in vitro. In the latter method, the hTR RNA and/or hTRT polypeptide may be purified before mixing.
  • the hTRT polypeptide is “purified” when it is separated from at least one other component of the in vitro expression system, and it may be purified to homogeneity as determined by standard methods (e.g., SDS-PAGE).
  • the in vitro reconstituted (IVR) telomerase has a variety of uses; in particular it is useful for identifying agents that modulate hTRT activity (e.g., drug screening assays).
  • the invention provides hTRT deletion variants that lack telomerase catalytic activity (i.e., having less than 1% of the wild type activity), as well as polynucleotides encoding the variants lacking telomerase catalytic activity.
  • the invention provides variants comprising one or more of the following deletions relative to wild-type hTRT: residues 192-450, 637-660, 638-660, 748-766, 748-764, and 1055-1071. These variants are referred to herein as “PCA ⁇ variants” (processive telomerase catalytic activity minus variants).
  • PCA ⁇ variant proteins and polynucleotides of the invention lacking telomerase catalytic activity are used in, inter alia, therapeutic, screening and other applications.
  • PCA ⁇ variants are useful as dominant negative mutants for inhibition of telomerase activity in a cell.
  • a PCA ⁇ variant is introduced into a cell (e.g., by transfection with a polynucleotide expression vector expressing the PCA ⁇ variant), resulting in sequestration of a cell component (e.g., hTR) required for accurate telomere elongation.
  • a cell component e.g., hTR
  • telomere extension e.g., by displacing active telomerase from telomeric DNA.
  • a PCA ⁇ variant of the invention having one or several hTRT activities i.e., other than processive telomerase catalytic activity
  • the hTRT variant polypeptides and polynucleotides of the invention may be produced using any of a variety of techniques known in the art.
  • a polypeptide having the desired sequence, or a polynucleotide encoding the polypeptide is chemically synthesized (see, e.g., Roberge, et al., 1995, Science 269:202; Brown et al., 1979, Meth. Enzymol. 68:109). More often, the hTRT variant polypeptides and polynucleotides of the invention are created by manipulation of a recombinant polynucleotide encoding an hTRT polypeptide. Examples of suitable recombinant polynucleotides include pGRN121, supra, and other hTRT cDNA and genomic sequences.
  • One convenient method for producing a polynucleotide encoding a desired hTRT deletion variant is by restriction digestion and subsequent ligation of a hTRT polynucleotide, to remove a region(s) of the polynucleotide encoding the amino acid residues to be deleted. If desired, restriction sites can be introduced or removed from a synthetic or naturally occurring hTRT gene to facilitate the production and detection of variants.
  • the recombinant polynucleotide encoding an hTRT variant of the invention is linked to appropriate regulatory elements (e.g., promoters, enhancers, polyadenylation signals, and the like) and expressed in a cell free system (see, e.g., Weinrich et al., supra), in bacteria (e.g., E. coli ), in ex vivo animal cell culture (see, e.g., Bodnar et al., supra), in animals or plants (e.g., transgenic organisms or in gene therapy applications), or by any other suitable method.
  • appropriate regulatory elements e.g., promoters, enhancers, polyadenylation signals, and the like
  • Suitable expression systems are well known in the art and include those described in Weinrich et al., and Bodnar et al., both supra, and in e.g., copending U.S. patent application Ser. No. 08/912,951, still pending and Ser. No. 08/974,549, now U.S. Pat. No. 6,166,178.
  • Additional hTRT variants of the invention may be made using “DNA shuffling” in vitro recombination technology (see, e.g., Crameri et al., 1998, Nature 391:288-291; Patten et al., 1997, Curr. Opin. Biotechnol.
  • wild-type hTRT and non-human hTRT-homologs may be used in individually or various combinations as starting substrates to produce novel polypeptides with the desired activity.
  • the activity or activities of the resulting polypeptides determined using the assays described in Section I, supra.
  • Exemplary hTRT variants were created by in vitro mutagenesis of polynucleotides encoding a full-length hTRT protein using the method of Perez et al., 1994, J. Biol. Chem. 269:22485-87.
  • the mutant polynucleotides were expressed in vitro and telomerase reconstituted by in vitro mixing of hTRT and human telomerase RNA as described in Weinrich et al., 1997, supra. Reconstitution reactions were carried out using 0.5 pmole, 20 pmole, and, in some cases, other amounts of added hTR.
  • Telomerase processive catalytic activity was assayed using a modified TRAP assay (Weinrich et al., 1997, supra). The results are summarized in Table 1, infra.
  • hTRT variants described supra are altered in their ability to bind hTR.
  • the variants encoded by pGRN235, pGRN242 and pGRN243 exhibited telomerase activity when 20 pmoles hTR (template RNA) was included in the reconstitution reaction, but showed a low or undetectable level of activity when 0.5 pmoles of hTR was used.
  • the variable activity of these variants indicates that these variants have altered (e.g., decreased) hTR binding activity.
  • the region from 415 to 450 is likely involved in RNA binding (e.g., by affecting the conformation of the protein).
  • hTR binding to hTRT was also affected by mutations and deletions in the region 560-565.
  • RNA binding was assayed by adding purified hTR to epitope tagged TRT proteins (i.e., including a FLAG sequence; Immunex Corp, Seattle Wash.). The hTR and protein were incubated under conditions under which tagged “wild-type” hTRT associates with template RNA (hTR), and the hTRT protein or hTRT-hTR complex (if present) were immunoprecipitated. The precipitated complex was assayed for the presence and amount of associated RNA.
  • telomerase reconstitution may be inhibited using peptides comprising the sequence corresponding the hTRT residues 405-418, 560-565, or fragments thereof, or peptide mimetics of such sequences.
  • telomerase activity in a cell or an in vitro composition in which TRT protein and TR RNA are present is reduced by introducing to the cell or in vitro composition a polypeptide comprising the sequence FFYVTE (SEQ ID NO:3), a polypeptide comprising the sequence YGVLLKTHCPLRAA(SEQ ID NO:4), a polypeptide consisting essentially of FFYVTE(SEQ ID NO:3), a polypeptide consisting essentially of FYVT(SEQ ID NO:5), a polypeptide consisting essentially of YGVLLKTHCPLRAA(SEQ ID NO:4), a fragment of at least three residues of the aforementioned polypeptides, or a peptide analog or mimetic of the polypeptide of any of the aforementioned compositions.
  • Peptide mimetics are well known and are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template polypeptide (Fauchere, 1986, Adv. Drug Res. 15:29; Veber et al., 1985, TINS p. 392; and Evans et al., 1987, J. Med. Chem. 30:1229).
  • peptidomimetics are structurally similar to the paradigm polypeptide having the sequence from hTRT but have one or more peptide linkages optionally replaced by a linkage selected from the group consisting of: —CH2NH—, —CH2S—, —CH2—CH2—, —CH′CH— (cis and trans), —COCH2—, —CH(OH)CH2—, and —CH2SO—.
  • Peptide mimetics may have significant advantages over polypeptide embodiments of this invention, including, for example: more economical production, greater chemical stability, enhanced pharmacological properties (half-life, absorption, potency, efficacy, etc.), altered specificity (e.g., a broad-spectrum of biological activities), reduced antigenicity, and others.
  • synthetic or non-naturally occurring amino acids can also be used to substitute for the amino acids present in the polypeptide or in the functional moiety of fusion proteins.
  • Synthetic or non-naturally occurring amino acids refer to amino acids which do not naturally occur in vivo but which, nevertheless, can be incorporated into the peptide structures described herein.
  • Preferred synthetic amino acids are the d- ⁇ -amino acids of naturally occurring 1- ⁇ -amino acid, mentioned above, as well as non-naturally occurring d- and 1- ⁇ -amino acids represented by the formula H2NCHR5COOH where R5 is 1) a lower alkyl group, 2) a cycloalkyl group of from 3 to 7 carbon atoms, 3) a heterocycle of from 3 to 7 carbon atoms and 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur, and nitrogen, 4) an aromatic residue of from 6 to 10 carbon atoms optionally having from 1 to 3 substituents on the aromatic nucleus selected from the group consisting of hydroxyl, lower alkoxy, amino, and carboxyl, 5) -alkylene-Y where alkylene is an alkylene group of from 1 to 7 carbon atoms and Y is selected from the group consisting of (a) hydroxy, (b) amino, (c) cycloalkyl and cycloalkenyl of from 3 to 7
  • compositions e.g., polypeptides and mimetics
  • telomerase association and activity inhibitors other than the disclosed polypeptide and mimetics.
  • These compositions may be used, for example, in rational drug design for e.g., computer modeling of telomerase activity modulators (e.g., modulators that inhibit the association of TRT and TR or that catalyze the disassociation of the telomerase complex), as positive controls in screens for modulators of telomerase activity, or in competition assays with candidate telomerase activity modulators.
  • Mutagenesis of the hTRT coding sequence of pGRN125 was carried out using the methods described by Perez et al., 1994, J. Biol. Chem. 269:22485-87. Most of the deletion mutants were generated from the plasmid pGRN125 (Weinrich et al., 1997, supra). Deletion mutants pGRN235 and pGRN236 were made in a secondary round of mutagenesis in an altered pGRN234.
  • pGRN234 was generated by mutating (deleting) the Nco I site in pGRN125 (changing CAC to CAT in the histidine residue at position 754) and introducing a new NcoI site at the translation start site (ATG).
  • Table 2 shows exemplary oligonucleotides used to generate the plasmids expressing the deletion variants of the invention.
  • Oligo CCCTCAGAC creates a NheI site.
  • RT3A ACTCAGGCCCGGCCCCCGCCACACG 60 Mutagenesis oligo to create a deletion CTAGCGAGACCAAGCACTTCCTCTAC of aa 192-323 in pGRN125.
  • Oligo TCCTCAGGC creates a NheI site.
  • RT4A ACTCAGGCCCGGCCCCCGCCACACG 60 Mutagenesis oligo to create a deletion CTAGCGTGGTGTCACCTGCCAGACCC of aa 192-271 in pGRN125.
  • Oligo GCCGAAGAA creates a NheI site.
  • RT6A ATCCCCAAGCCTGACGGGCTGCGGC 69 Mutagenesis oligo to create a deletion CGATTGTTAACATGCTGTTCAGCGTG of aa 638-660 in pGRN125. Oligo CTCAACTACGAGCGGGCG creates a Hpa I site. RT8A ACGTACTGCGTGCGTCGGTATGCCGT 63 Mutagenesis oligo to create a deletion GGTCACAGATCTCCAGCCGTACATGC of aa 748-766 in pGRN125. Oligo GACAGTTCGTG creates a BgI II site.
  • RT3A/5 ACTCAGGCCCGGCCCCCGCCACACG 60 Mutagenesis oligo to create a deletion CTAGCCTGCTCCGCCAGCACAGCAG of aa 192-450 in pGRN125. Oligo CCCCTGGCAG creates a NheI site. LM121- GTTCAGATGCCGGCCCACGGCCTATT 63 Mutagenesis oligo to delete aa 930-934. WG CCCTCTAGATACCCGGACCCTGGAGG Oligo introduces a new XbaI site TGCAGAGCGAC LM122- CCCTGGGCCTGCCAGCCGGGTGC 50 Mutagenesis oligo to delete aa 222-240.
  • Nuc CGGCGCTGCCCCTGAGCCGGAGCGG Oligo introduces a new Nae I site RT3 GCTAGTGGACCCCGAAGGCGTCTGG 60 Mutagenesis oligo to create a deletion GATGCGAGACCAAGCACTTCCTCTAC of aa2OO-323 in pGRN125 TCCTCAGGC RT4 GCTAGTGGACCCCGAAGGCGTCTGG 60 Mutagenesis oligo to create a deletion GATGCGTGGTGTCACCTGCCAGACCC of aa 200-271 in pGRN125 GCCGAAGAA RT6 GACGGGCTGCGGCCGATTGTGAACA 60 Mutagenesis oligo to create a deletion TGGACCTGTTCAGCGTGCTCAACTAC of aa 638-660 in pGRN125 GAGCGGGCG RT8 ACGTACTGCGTGCGTCGGTATGCCGT 60 Mutagenesis oligo to create a deletion GGTCACCTTGACAGACCTCCAGCCGT of aa 748-764 in
  • a polypeptide region in a first polypeptide “corresponds” to a region in a second polypeptide when the amino acid sequences of the two regions, or flanking the two regions, are the same or substantially identical. Sequences can be aligned by inspection (e.g., alignment of identical sequences) or by computer implemented alignment of the two sequences. Thus, for example, the residues 192 to 323 of the hTRT polypeptide having the sequence set forth in FIG. 1 “correspond” to residues in the same position in a hTRT polypeptide that differs from the FIG. 1 sequence due to polymorphic variation, or other mutations or deletions (e.g., when the two polypeptides are optimally aligned).
  • Alignments may also be carried out using the GAP computer program, version 6.0 (Devereux et al, 1984, Nucl. Acid. Res. 12:387; available from the University of Wisconsin Genetics Computer Group, Madison, Wis.).
  • the GAP program utilizes the alignment method of Needleham and Wunsch, 1970 J. Mol. Biol. 48: 443-453 as revised by Smith and Waterman, 1981, Adv. Appl. Math 2:482.
  • the preferred default parameters for the GAP program include (1) the weighted comparison matrix of Gribskov and Burgess, 1986, Nuc. Acid. Res. 14:6745 as described by Schwartz and Dayhoff, eds., 1979, Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, pp.
  • 353-358 (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
  • a variant When referring to an “activity” of an hTRT variant, a variant is considered to be active in an assay of it displays at least 40% of the activity characteristic of the hTRT polypeptide having the sequence set forth in FIG. 1 (“wild type”). A variant is considered to lack activity when it has less that 1% of the “wild type” activity. A variant with greater than 1% activity and less than 40% activity has “intermediate activity.”
  • “conservative substitution,” refers to substitution of amino acids with other amino acids having similar properties (e.g. acidic, basic, positively or negatively charged, polar or non-polar).
  • the following six groups each contain amino acids that are conservative substitutions for one another: 1) alanine (A), serine (S), threonine (T); 2) aspartic acid (D), glutamic acid (E); 3) asparagine (N), glutamine (Q); 4) arginine (R), lysine (K); 5) isoleucine (I), leucine (L), methionine (M), valine (V); and 6) phenylalanine (F), tyrosine (Y), tryptophan (W) (see also, Creighton, 1984, Proteins, W. H. Freeman and Company).

Abstract

The invention provides compositions and methods related to human telomerase reverse transcriptase (hTRT), the catalytic protein subunit of human telomerase. Catalytically active human telomerase reverse transcriptase variants comprising deletions or other mutations are provided.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a continuation-in-part of U.S. patent application Ser. No. 09/052,864, filed Mar. 31, 1998, now abandoned.
The priority application is hereby incorporated herein by reference in its entirety, as are the following: U.S. application Ser. No. 08/851,843, filed May 6, 1997, now U.S. Pat. No. 6,093,809; Ser. No. 08/854,050, filed May 9, 1997, now U.S. Pat. No. 6,261,836; Ser. No. 08/911,312, filed Aug. 14, 1997, still pending; Ser. No. 08/912,951, filed Aug. 14, 1997, still pending; Ser. No. 08/915,503, filed Aug. 14, 1997, now abandoned; Ser. No. 08/974,549, filed Nov. 19, 1997, now U.S. Pat. No. 6,166,178; and Ser. No. 08/974,584, filed Nov. 18, 1997, still pending; and International Applications PCT/US97/17618 and PCT/US97/17885, which designate the U.S.
FIELD OF THE INVENTION
The present invention is related to the catalytic protein subunit of human telomerase. The invention provides methods and compositions relating to medicine, molecular biology, chemistry, pharmacology, and medical diagnostic and prognostic technology.
BACKGROUND OF THE INVENTION
The following discussion is intended to introduce the field of the present invention to the reader. The citation of various references in this section should not be construed as an admission of prior invention.
It has long been recognized that complete replication of the ends of eukaryotic chromosomes requires specialized cell components (Watson, 1972, Nature New Biol., 239:197; Olovnikov, 1973, J. Theor. Biol., 41:181). Replication of a linear DNA strand by conventional DNA polymerase requires an RNA primer, and can proceed only 5′ to 3′. When the RNA bound at the extreme 5′ ends of eukaryotic chromosomal DNA strands is removed, a gap is introduced, leading to a progressive shortening of daughter strands with each round of replication. This shortening of telomeres, the protein-DNA structures physically located on the ends of chromosomes, is thought to account for the phenomenon of cellular senescence or aging of normal human somatic cells in vitro and in vivo. The maintenance of telomeres is a function of a telomere-specific DNA polymerase known as telomerase. Telomerase is a ribonucleoprotein (RNP) that uses a portion of its RNA moiety as a template for telomeric DNA synthesis (Morin, 1997, Eur. J. Cancer 33:750). The length and integrity of telomeres and the telomerase expression status of a cell is thus related to entry of a cell into a senescent stage (i.e., loss of proliferative capacity), or the ability of a cell to escape senescence, i.e., to become immortal.
Consistent with the relationship of telomeres and telomerase to the proliferative capacity of a cell (i.e., the ability of the cell to divide indefinitely), telomerase activity is detected in immortal cell lines and an extraordinarily diverse set of tumor tissues, but is not detected (i.e., was absent or below the assay threshold) in normal somatic cell cultures or normal tissues adjacent to a tumor (see, U.S. Pat. Nos. 5,629,154; 5,489,508; 5,648,215; and 5,639,613; see also, Morin, 1989, Cell 59: 521; Shay and Bacchetti 1997, Eur. J. Cancer 33:787; Kim et al., 1994, Science 266:2011; Counter et al., 1992, EMBO J. 11:1921; Counter et al., 1994, Proc. Natl. Acad. Sci. U.S.A. 91, 2900; Counter et al., 1994, J. Virol. 68:3410). Moreover, a correlation between the level of telomerase activity in a tumor and the likely clinical outcome of the patient has been reported (e.g., U.S. Pat. No. 5,639,613, supra; Langford et al., 1997, Hum. Pathol. 28:416). Thus, human telomerase is an ideal target for diagnosing and treating human diseases relating to cellular proliferation and senescence, such as cancer, or for increasing the proliferative capacity of a cell.
BRIEF SUMMARY OF THE INVENTION
In one aspect, the invention provides an isolated or recombinant hTRT polypeptide that has telomerase catalytic activity. In one embodiment, the hTRT polypeptide has a deletion of at least 25 residues in the regions corresponding to residues 192-323, 200-323, 192-271, 200-271, 222-240, 415-450, 192-323 and 415-450, or 192-271 and 415-450 of hTRT. In a related embodiment, residues 192-323, 200-323, 192-271, 200-271, 222-240, 415-450, 192-323 and 415-450, or 192-271 and 415-450 of hTRT are deleted. The invention also provides a polynucleotide comprising a nucleotide sequence encoding these hTRT polypeptides. In some embodiments, the polynucleotide includes a promoter sequence operably linked to the nucleotide sequence encoding the hTRT polypeptide.
The invention also provides a method of preparing recombinant telomerase by contacting a recombinant hTRT polypeptide containing a deletion as described supra with a telomerase RNA component under conditions such that the recombinant protein and the telomerase RNA component associate to form a telomerase enzyme capable of catalyzing the addition of nucleotides to a telomerase substrate. The hTRT polypeptide may be produced in an in vitro expression system and/or may be purified before the contacting step. In some embodiments, the contacting occurs in a cell.
The invention further provides a method for increasing the proliferative capacity of a vertebrate cell by introducing into a cell the recombinant hTRT polynucleotide encoding an hTRT deletion variant described supra. In a related embodiment, the invention provides a cell, such as a human cell or other mammalian cell, comprising a nucleotide sequence that encodes the hTRT deletion variant polypeptide. The invention provides such cells that have an increased proliferative capacity relative to a cell that is otherwise identical but does not comprise the recombinant polynucleotide.
In a different aspect of the invention, an isolated or recombinant hTRT polypeptide that has a deletion of amino acid residues 192-450, 560-565, 637-660, 638-660, 748-766, 748-764, or 1055-1071, where the residue numbering is with reference to the hTRT polypeptide having the sequence provided in FIG. 1, is provided. In a related aspect, the invention provides an isolated, recombinant, or substantially purified polynucleotide encoding this polypeptide, which in some embodiments includes a promoter sequence operably linked to the nucleotide sequence encoding the hTRT polypeptide.
The invention also provides a method of reducing telomerase activity in a cell by introducing the polynucleotide described supra (i.e., having a deletion of deletion of amino acid residues 192-450, 560-565, 637-660, 638-660, 748-766, 748-764, or 1055-1071) into a cell under conditions in which it is expressed.
In a related embodiment, the hTRT polypeptide has one or more mutations other than, or in addition to, a deletion of at least 25 residues in the regions corresponding to residues 192-323, 200-323, 192-271, 200-271, 222-240, 415-450, 192-323 and 415-450, or 192-271 and 415-450 of hTRT.
DESCRIPTION OF THE FIGURES
FIG. 1 shows the amino acid sequence of a 1132-residue human telomerase reverse transcriptase (hTRT) protein (SEQ ID NO:2).
FIG. 2 shows the nucleotide sequence of a naturally occurring cDNA encoding the hTRT protein (SEQ ID NO:1).
DETAILED DESCRIPTION OF THE INVENTION
I. Introduction
Telomerase is a ribonucleoprotein complex (RNP) comprising an RNA component and a catalytic protein component. The catalytic protein component of human telomerase, hereinafter referred to as telomerase reverse transcriptase (“hTRT”), has been cloned, and protein, cDNA and genomic sequences determined. See, e.g., Nakamura et al., 1997, Science 277:955, and copending U.S. patent application Ser. No. 08/912,951 filed Aug. 14, 1997, still pending and Ser. No. 08/974,549, filed Nov. 19, 1997, now U.S. Pat. No. 6,166,178. The sequence of a full-length native hTRT has been deposited in GenBank (Accession No. AF015950), and plasmid and phage vectors having hTRT coding sequences have been deposited with the American Type Culture Collection, Rockville, Md. (accession numbers 209024, 209016, and 98505). The catalytic subunit protein of human telomerase has also been referred to as “hEST2” (Meyerson et al., 1997, Cell 90:785), “hTCS1” (Kilian et al., 1997, Hum. Mol. Genet. 6:2011), “TP2” (Harrington et al., 1997, Genes Dev. 11:3109), and “hTERT” (e.g., Greider, 1998, Curr. Biol. 8:R178-R181). Human TRT is also described in the aforereferenced priority applications and U.S. patent application Ser. No. 08/846,017, now abandoned, Ser. No. 08/844,419, now abandoned, and Ser. No. 08/724,643,now abandoned. The RNA component of human telomerase (hTR) has also been characterized (see U.S. Pat. No. 5,583,016). All of the aforementioned applications and publications are incorporated by reference herein in their entirety and for all purposes.
Human TRT is of extraordinary interest and value because, inter alia, telomerase activity in human cells and other mammalian cells correlates with cell proliferative capacity, cell immortality, and the development of a neoplastic phenotype. Thus, hTRT polypeptides, including the hTRT variants described herein, and polynucleotides encoding hTRT polypeptides, are used, inter alia for conferring a telomerase activity (e.g., telomerase catalytic activity, infra) in a telomerase-negative cell such as a cell from a human, a mammal, a vertebrate, or other eukaryote ( see, e.g., Bodnar et al., 1998, Science 279:349 and copending U.S. patent application Ser. No. 08/912,951, still pending and Ser. No. 08/974,549, now U.S. Pat. No. 6,166,178). Variants that lack at least one hTRT activity (e.g., telomerase catalytic activity) are used, inter alia, to inhibit telomerase activity in a cell (e.g., by acting as “dominant negative mutants”). The hTRT variants and polynucleotides encoding them, as described herein, are similarly useful in screening assays for identifying agents that modulate telomerase activity.
The hTRT variants of the present invention are characterized by one or more deletions or mutations, relative to a naturally occurring hTRT polypeptide, in defined regions of the protein, as described in detail infra. These hTRT variants may have none, one, or several of the biological activities that may be found in naturally-occurring full-length hTRT proteins. These activities include telomerase catalytic activity (the ability to extend a DNA primer that functions as a telomerase substrate by adding a partial, one, or more than one repeat of a sequence, e.g., TTAGGG, encoded by a template nucleic acid, e.g., hTR), telomerase conventional reverse transcriptase activity (see Morin, 1997, supra, and Spence et al., 1995, Science 267:988); nucleolytic activity (see Morin, 1997, supra; Collins and Grieder, 1993, Genes and Development 7:1364; Joyce and Steitz, 1987, Trends Biochem. Sci. 12:288); primer (telomere) binding activity (see, Morin, 1997, supra; Collins et al., 1995, Cell 81:677; Harrington et al., 1995, J. Biol. Chem. 270:8893); dNTP binding activity (Morin, 1997, supra; Spence et al., supra); and RNA (e.g.. hTRT) binding activity (see Morin, 1997, supra; Harrington et al., 1997, Science 275:973; Collins et al., 1995, Cell 81:677).
In one embodiment of the invention, the hTRT variant has telomerase catalytic activity. Telomerase catalytic activity may be processive or nonprocessive. Processive telomerase catalytic activity occurs when a telomerase RNP adds multiple repeats to a primer or telomerase before the DNA is released by the enzyme complex (see, e.g., Morin, 1989, Cell 59:521 and Morin, 1997, Eur. J. Cancer 33:750). Nonprocessive activity occurs when telomerase adds a partial, or only one, repeat to a primer and is then released (see Morin, 1997, supra). In a particular embodiment of the invention, the hTRT variant has processive telomerase catalytic activity.
Processive telomerase catalytic activity can be assayed by a variety of methods, including the “conventional assay” (Morin, 1989, Cell 59:521), the TRAP assay (U.S. Pat. No. 5,629,154; see also, PCT publication WO 97/15687, PCT publication WO 95/13381; Krupp et al. Nucleic Acids Res., 1997, 25: 919; Wright et al., 1995, Nuc. Acids Res. 23:3794), the “dot blot immunoassay” (U.S. patent application Ser. No. 08/833,377 now U.S. Pat. No. 5,968,506), and other assays (e.g., Tatematsu et al., 1996, Oncogene 13:2265). The TRAPeze™ Kit (Oncor, Inc., Gaithersburg, Md.) may be used. The telomerase substrate used in these assays may have a natural telomere sequence, or may be have a synthetic oligonucleotide with a different sequence (see, e.g., Morin, 1989, Cell 59:521; Morin, 1991, Nature 353:454-56).
As used herein, an hTRT variant is considered to have a specified activity if the activity is exhibited by either the hTRT variant polypeptide without an associated hTR RNA or in an hTRT-hTR complex. Each of the hTRT activities described supra is also described in detail in copending U.S. patent applications Ser. No. 08/912,951 still pending and 08/974,549, now U.S. Pat. No. 6,166,178.
II. hTRT Variants Described
a) hTRT Variants with Telomerase Catalytic Activity
It has now been demonstrated that large regions of the hTRT protein can be mutated (e.g., deleted) without loss of telomerase catalytic activity. Sites of mutation (e.g., deletion) are described herein with reference to the amino acid sequence provided in FIG. 1 and encoded in plasmid pGRN121 (ATCC accession number 209016); however it will be recognized that the same or equivalent mutations may be made in other hTRT polypeptides, e.g., naturally occurring variants such as polymorphic variants, hTRT fusion proteins, hTRT homologs (e.g., from non-human species), and the like. For ease of discussion, the residues of the full-length hTRT protein having a sequence as provided in FIG. 1 are referred to herein by number, with the amino-terminal methionine (M) in FIG. 1 numbered “1”, and the carboxy-terminal aspartic acid (D) numbered “1132”.
Regions of the hTRT protein that can be mutated (e.g., deleted) without abolishing telomerase catalytic activity include the regions from amino acid residues 192 to 323 (inclusive) and residues 415 to 450 (inclusive). As is demonstrated in the experiments described infra, all or part of either of these regions, or all or part of both of them, can be deleted without abolishing the telomerase catalytic activity of the protein. The regions from amino acid residues 192 to 323 and residues 415 to 450 may be referred to as “nonessential” regions of hTRT (i.e., not essential for telomerase catalytic activity). Thus, in various embodiments, the hTRT variants of the invention comprise deletions of, or other mutations in, these nonessential regions of hTRT. As described in Section IV, infra, certain mutations (e.g., deletion of residues 415-450) alter RNA-binding characteristics of the hTRT variant.
Examples of mutations that can be made in the hTRT polypeptides of the invention include deletions, insertions, substitutions, and combination of mutations. Thus, in some embodiments the mutation is a deletion of at least one, typically at least about 10, and often at least about 25, at least about 50, or at least about 100 amino acid residues relative to a naturally occurring hTRT. In alternative embodiments, the mutation is a single amino acid substitution in a “non-essential” region, or a combinations of substitutions. Substitutions may be conservative substitutions or non-conservative substitutions. In still other embodiments, the mutation is an insertion or substitution of amino acids, for example the insertion of residues that encode an epitope tag or novel proteolytic site. Substitutions may be of one or more (e.g., all) of the residues in the above-mentioned regions or may be combined with deletions so that, e.g., a shorter heterologous sequence is a substituted for a longer hTRT sequence. It will be appreciated, as noted supra, that in some embodiments the hTRT variant has more than one different type of mutation relative to a naturally occurring hTRT protein (e.g., a deletion and a point mutation).
The hTRT variants of the invention have certain advantages compared to naturally occurring hTRT proteins. In some embodiments, mutations may confer more efficient in vitro expression of active hTRT (e.g., in expression systems in which shorter polypeptides are more efficiently expressed than longer polypeptides), may provide sequences that aid in purification (e.g., an epitope tag sequence), or may add a new functional moiety to the hTRT polypeptide (e.g., a 3′→5′ exonuclease domain from DNA polymerase I).
As noted supra, the hTRT variant polypeptides of the invention comprising mutations (e.g., deletions) in the “non-essential” regions of the hTRT retain telomerase catalytic activity. These variants, and polynucleotides that encode them, are useful in any application for which other catalytically active hTRT proteins (e.g., wild-type hTRT proteins) or polynucleotides may be used, including, inter alia, in therapeutic, diagnostic, and screening uses. Exemplary uses of hTRT polypeptides and polynucleotides are described in additional detail in the afore cited copending applications (e.g., U.S. Ser. No. 08/912,951, still pending and Ser. No. 08/974,549,now U.S. Pat. No. 6,166,178).
In one embodiment, the hTRT variant of the invention is used to increase the proliferative capacity of a cell by, e.g., increasing telomerase activity in the cell (see, Bodnar et al. supra, and copending U.S. patent application Ser. No. 08/912,951, still pending and Ser. No. 08/974,549, now U.S. Pat. No. 6,166,178 for a detailed description of exemplary methods). Briefly, in one embodiment, a polynucleotide comprising (i) a sequence encoding the hTRT variant polypeptide; (ii) an operably linked promoter (e.g., a heterologous promoter); and, (iii) optionally polyadenylation and termination signals, enhancers, or other regulatory elements, is introduced into a target cell (e.g., by transfection, lipofection, electroporation, or any other suitable method) under conditions in which the hTRT variant polypeptide is expressed. The expression in the cell of the catalytically active hTRT variant of the invention results in increased proliferative capacity (e.g., an immortal phenotype).
In another embodiment, the hTRT variant is used for in vitro reconstitution (IVR) of a telomerase ribonucleoprotein (e.g., comprising the hTRT variant polypeptide and a template RNA, e.g., hTR) that has telomerase catalytic activity. In vitro reconstitution methods are described in, e.g., Weinrich et al., 1997, Nat. Genet. 17:498, and copending U.S. patent application Ser. No. 08/912,951, still pending and Ser. No. 08/974,549, now U.S. Pat. No. 6,166,178. Briefly, in one embodiment, an expression vector encoding an hTRT variant of the invention is expressed in an in vitro expression system (e.g., a coupled transcription-translation reticulocyte lysate system such as that described in U.S. Pat. No. 5,324,637). In a particular embodiment, the hTRT variant polypeptide is coexpressed with hTR. In an alternative embodiment, the hTRT variant and hTR are separately expressed and then combined (mixed) in vitro. In the latter method, the hTR RNA and/or hTRT polypeptide may be purified before mixing. In this context, the hTRT polypeptide is “purified” when it is separated from at least one other component of the in vitro expression system, and it may be purified to homogeneity as determined by standard methods (e.g., SDS-PAGE). The in vitro reconstituted (IVR) telomerase has a variety of uses; in particular it is useful for identifying agents that modulate hTRT activity (e.g., drug screening assays).
(b) Deletion Variants Lacking Telomerase Catalytic Activity
In an other aspect, the invention provides hTRT deletion variants that lack telomerase catalytic activity (i.e., having less than 1% of the wild type activity), as well as polynucleotides encoding the variants lacking telomerase catalytic activity. In particular, the invention provides variants comprising one or more of the following deletions relative to wild-type hTRT: residues 192-450, 637-660, 638-660, 748-766, 748-764, and 1055-1071. These variants are referred to herein as “PCA variants” (processive telomerase catalytic activity minus variants).
The PCA variant proteins and polynucleotides of the invention lacking telomerase catalytic activity are used in, inter alia, therapeutic, screening and other applications. For example, PCA variants are useful as dominant negative mutants for inhibition of telomerase activity in a cell. In one embodiment, a PCA variant is introduced into a cell (e.g., by transfection with a polynucleotide expression vector expressing the PCA variant), resulting in sequestration of a cell component (e.g., hTR) required for accurate telomere elongation. Thus, for example, administration of a polypeptide that binds hTR, a DNA primer, a telomerase-associated protein, or other cell component, but which does not have telomerase catalytic activity, is used to reduce endogenous telomerase activity in the cell or to otherwise interfere with telomere extension (e.g., by displacing active telomerase from telomeric DNA). Similarly, in certain embodiments, a PCA variant of the invention having one or several hTRT activities (i.e., other than processive telomerase catalytic activity) is used for screening for agents that specifically modulate (inhibit or activate) a telomerase activity other than telomerase catalytic activity. The use of hTRT variants as dominant negative mutants, and in other applications, is described in detail in copending U.S. patent application Ser. No. 08/912,951, still pending and Ser. No. 08/974,549, now U.S. Pat. No. 6,166,178.
III. Making hTRT Variants
The hTRT variant polypeptides and polynucleotides of the invention may be produced using any of a variety of techniques known in the art. In one embodiment, a polypeptide having the desired sequence, or a polynucleotide encoding the polypeptide, is chemically synthesized (see, e.g., Roberge, et al., 1995, Science 269:202; Brown et al., 1979, Meth. Enzymol. 68:109). More often, the hTRT variant polypeptides and polynucleotides of the invention are created by manipulation of a recombinant polynucleotide encoding an hTRT polypeptide. Examples of suitable recombinant polynucleotides include pGRN121, supra, and other hTRT cDNA and genomic sequences.
Methods for cloning and manipulation of hTRT encoding nucleic acids (e.g., site-specific mutagenesis, linker scanning mutagenesis, and the like) are well known in the art and are described, for example, in Sambrook et al., 1989, Molecular Cloning: a Laboratory Manual, 2nd Ed., Vols. 1-3, Cold Spring Harbor Laboratory, and Ausubel et al., 1997, Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience, New York. One convenient method for producing a polynucleotide encoding a desired hTRT deletion variant is by restriction digestion and subsequent ligation of a hTRT polynucleotide, to remove a region(s) of the polynucleotide encoding the amino acid residues to be deleted. If desired, restriction sites can be introduced or removed from a synthetic or naturally occurring hTRT gene to facilitate the production and detection of variants.
Typically, the recombinant polynucleotide encoding an hTRT variant of the invention is linked to appropriate regulatory elements (e.g., promoters, enhancers, polyadenylation signals, and the like) and expressed in a cell free system (see, e.g., Weinrich et al., supra), in bacteria (e.g., E. coli), in ex vivo animal cell culture (see, e.g., Bodnar et al., supra), in animals or plants (e.g., transgenic organisms or in gene therapy applications), or by any other suitable method. Suitable expression systems are well known in the art and include those described in Weinrich et al., and Bodnar et al., both supra, and in e.g., copending U.S. patent application Ser. No. 08/912,951, still pending and Ser. No. 08/974,549, now U.S. Pat. No. 6,166,178.
Additional hTRT variants of the invention may be made using “DNA shuffling” in vitro recombination technology (see, e.g., Crameri et al., 1998, Nature 391:288-291; Patten et al., 1997, Curr. Opin. Biotechnol. 8:724-733, Stemmer, 1994, Nature 370:389-391; Crameri et al., 1996, Nature Medicine, 2:1-3; Crameri et al., 1996, Nature Biotechnology 14:315-319; WO 95/22625; Stemmer, 1995, Science 270:1510; Stemmer et al., 1995, Gene, 164, 49-53; Stemmer, 1995, Bio/Technology, 13:549-553; Stemmer, 1994, Proc. Natl. Acad. Sci. USA 91:10747-10751). The specific deletion variants described supra, “wild-type hTRT” and non-human hTRT-homologs may be used in individually or various combinations as starting substrates to produce novel polypeptides with the desired activity. The activity or activities of the resulting polypeptides determined using the assays described in Section I, supra.
IV. Exemplary hTRT Variants
a) Generally
Exemplary hTRT variants were created by in vitro mutagenesis of polynucleotides encoding a full-length hTRT protein using the method of Perez et al., 1994, J. Biol. Chem. 269:22485-87. The mutant polynucleotides were expressed in vitro and telomerase reconstituted by in vitro mixing of hTRT and human telomerase RNA as described in Weinrich et al., 1997, supra. Reconstitution reactions were carried out using 0.5 pmole, 20 pmole, and, in some cases, other amounts of added hTR. Telomerase processive catalytic activity was assayed using a modified TRAP assay (Weinrich et al., 1997, supra). The results are summarized in Table 1, infra.
TABLE 1
Deletion Name Oligo Amino acids deleted Activity1
pGRN234 RT1 + RT2 none (delete NcoI site) +
pGRN226 RT3A 192-323 +
RT3 RT3 200-326 +
pGRN237 RT4A 192-271 +
RT4 RT4 200-271 +
pGRN210 LM122-Nuc 222-240 +
pGRN235 RT5 415-450 +
pGRN242 RT3A + RT5 192-326 + 415-450 +
pGRN243 RT4A + RT5 192-271 + 415-450 +
pGRN240 RT3A/5 192-450
pGRN238 RT6A 637-660
RT6 RT 6 638-660
pGRN239 RT8A 748-766
RT8 RT8 748-764
pGRN241 RT10 1055-1071
pGRN236 RT11 1084-1116
pGRN209 LM121-WG 930-934
pGRN231 560-565
1“+” = at least 40% activity compared to in vitro reconsitution using wild-type hTRT (e.g., encoded by pGRN125; see Weinrich et al., 1997, supra); “−” = less than 1% activity.
Certain of the hTRT variants described supra are altered in their ability to bind hTR. The variants encoded by pGRN235, pGRN242 and pGRN243 exhibited telomerase activity when 20 pmoles hTR (template RNA) was included in the reconstitution reaction, but showed a low or undetectable level of activity when 0.5 pmoles of hTR was used. The variable activity of these variants indicates that these variants have altered (e.g., decreased) hTR binding activity. Thus, the region from 415 to 450 is likely involved in RNA binding (e.g., by affecting the conformation of the protein).
This result suggests that the region immediately upstream of residue 415, corresponding to the conserved “CP” domain (Bryan et al., 1998, Proc. Nat'l. Acad. Sci. 95:8479-8484) is a region of contact between the hTRT protein and hTR (e.g., corresponding to about residues 405 to 418 as set forth in FIG. 1). This conclusion is supported by the relative lack of conservation of sequence when human and mouse TRT sequences are compared in the region corresponding tohTRT residues 415-450.
hTR binding to hTRT was also affected by mutations and deletions in the region 560-565. RNA binding was assayed by adding purified hTR to epitope tagged TRT proteins (i.e., including a FLAG sequence; Immunex Corp, Seattle Wash.). The hTR and protein were incubated under conditions under which tagged “wild-type” hTRT associates with template RNA (hTR), and the hTRT protein or hTRT-hTR complex (if present) were immunoprecipitated. The precipitated complex was assayed for the presence and amount of associated RNA. Deletion of residues 560-565 dramatically decreased the binding of hTR by hTRT, with the concurrent expected decrease in telomerase activity (see Table 1, pGRN231). Mutation of phenylalanine (F) to alanine (A) mutation at position 561 of hTRT (the “F561A” variant; see, Weinrich et al., 1997, supra) resulted in reduced binding of hTR: this variant did not effectively bind hTR in association reactions when hTR was present at 0.5 pmoles, and showed less-than wild-type binding at 20 pmoles hTR. Mutation of tyrosine at 562 to alanine similarly resulted in a loss of hTR binding activity (e.g., about a 70-80% reduction compared to the wild-type sequence). Mutation of threonine at position 564 to alanine resulted in a decrease in RNA binding by approximately 20% compared to wild-type. In contrast, mutation of residues 560 (F) and 565 (E) to alanine did not affect hTR binding. These results indicate that the region from 560-565 is involved in RNA binding, e.g., by providing residues that contact hTR.
As will be apparent to one of skill advised of these results, the telomerase reconstitution may be inhibited using peptides comprising the sequence corresponding the hTRT residues 405-418, 560-565, or fragments thereof, or peptide mimetics of such sequences. Thus, in one embodiment of the present invention, telomerase activity in a cell or an in vitro composition in which TRT protein and TR RNA are present, such as a telomerase reconstitution assay, is reduced by introducing to the cell or in vitro composition a polypeptide comprising the sequence FFYVTE (SEQ ID NO:3), a polypeptide comprising the sequence YGVLLKTHCPLRAA(SEQ ID NO:4), a polypeptide consisting essentially of FFYVTE(SEQ ID NO:3), a polypeptide consisting essentially of FYVT(SEQ ID NO:5), a polypeptide consisting essentially of YGVLLKTHCPLRAA(SEQ ID NO:4), a fragment of at least three residues of the aforementioned polypeptides, or a peptide analog or mimetic of the polypeptide of any of the aforementioned compositions.
Peptide mimetics (or peptide analogs) are well known and are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template polypeptide (Fauchere, 1986, Adv. Drug Res. 15:29; Veber et al., 1985, TINS p.392; and Evans et al., 1987, J. Med. Chem. 30:1229). Generally, peptidomimetics are structurally similar to the paradigm polypeptide having the sequence from hTRT but have one or more peptide linkages optionally replaced by a linkage selected from the group consisting of: —CH2NH—, —CH2S—, —CH2—CH2—, —CH′CH— (cis and trans), —COCH2—, —CH(OH)CH2—, and —CH2SO—. Peptide mimetics may have significant advantages over polypeptide embodiments of this invention, including, for example: more economical production, greater chemical stability, enhanced pharmacological properties (half-life, absorption, potency, efficacy, etc.), altered specificity (e.g., a broad-spectrum of biological activities), reduced antigenicity, and others. In addition to modifications to the peptide backbone, synthetic or non-naturally occurring amino acids can also be used to substitute for the amino acids present in the polypeptide or in the functional moiety of fusion proteins. Synthetic or non-naturally occurring amino acids refer to amino acids which do not naturally occur in vivo but which, nevertheless, can be incorporated into the peptide structures described herein. Preferred synthetic amino acids are the d-α-amino acids of naturally occurring 1-α-amino acid, mentioned above, as well as non-naturally occurring d- and 1-α-amino acids represented by the formula H2NCHR5COOH where R5 is 1) a lower alkyl group, 2) a cycloalkyl group of from 3 to 7 carbon atoms, 3) a heterocycle of from 3 to 7 carbon atoms and 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur, and nitrogen, 4) an aromatic residue of from 6 to 10 carbon atoms optionally having from 1 to 3 substituents on the aromatic nucleus selected from the group consisting of hydroxyl, lower alkoxy, amino, and carboxyl, 5) -alkylene-Y where alkylene is an alkylene group of from 1 to 7 carbon atoms and Y is selected from the group consisting of (a) hydroxy, (b) amino, (c) cycloalkyl and cycloalkenyl of from 3 to 7 carbon atoms, (d) aryl of from 6 to 10 carbon atoms optionally having from 1 to 3 substituents on the aromatic nucleus selected from the group consisting of hydroxyl, lower alkoxy, amino and carboxyl, (e) heterocyclic of from 3 to 7 carbon atoms and 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur, and nitrogen, (f) —C(O)R2 where R2 is selected from the group consisting of hydrogen, hydroxy, lower alkyl, lower alkoxy, and —NR3R4 where R3 and R4 are independently selected from the group consisting of hydrogen and lower alkyl, (g) —S(O)nR6 where n is an integer from 1 to 2 and R6 is lower alkyl and with the proviso that R5 does not define a side chain of a naturally occurring amino acid. Other preferred synthetic amino acids include amino acids wherein the amino group is separated from the carboxyl group by more than one carbon atom such as β-alanine, γ-aminobutyric acid, and the like.
It will also be recognized by those of skill upon reviewing these results that the compositions (e.g., polypeptides and mimetics) described supra can be used to identify telomerase association and activity inhibitors other than the disclosed polypeptide and mimetics. These compositions may be used, for example, in rational drug design for e.g., computer modeling of telomerase activity modulators (e.g., modulators that inhibit the association of TRT and TR or that catalyze the disassociation of the telomerase complex), as positive controls in screens for modulators of telomerase activity, or in competition assays with candidate telomerase activity modulators.
b) Methods
1) Mutagenesis
Mutagenesis of the hTRT coding sequence of pGRN125 was carried out using the methods described by Perez et al., 1994, J. Biol. Chem. 269:22485-87. Most of the deletion mutants were generated from the plasmid pGRN125 (Weinrich et al., 1997, supra). Deletion mutants pGRN235 and pGRN236 were made in a secondary round of mutagenesis in an altered pGRN234. pGRN234 was generated by mutating (deleting) the Nco I site in pGRN125 (changing CAC to CAT in the histidine residue at position 754) and introducing a new NcoI site at the translation start site (ATG). Table 2 shows exemplary oligonucleotides used to generate the plasmids expressing the deletion variants of the invention.
TABLE 2
Oligo
Name Oligo sequence 5′-3′ length Description
RT1 GAAGGCCGCCCACGGGCACGTCCGC 25 Mutagenesis oligo to delete Nco 1 site
from pGRNI25
RT2 CCCGGCCACCCCAGCCATGGCGCGC 32 Mutagenesis oligo to create Nco 1 site
GCTCCCC @ATG of pGRN 125
RT5 TACGGGGTGCTCCTCAAGACGCACTG 60 Mutagenesis oligo to create a deletion
CCCGCTGCTCCGCCAGCACAGCAGC of aa 415-450 in pGRNl25
CCCTGGCAG
RT10 TACTCCATCCTGAAAGCCAAGAACGC 60 Mutagenesis oligo to create a deletion
AGGGCTGTGCCACCAAGCATTCCTGC of aa 1055-1071 in pGRN125
TCAAGCTG
RT11 CTGTGCCACCAAGCATTCCTGCTCAA 60 Mutagenesis oligo to create a deletion
GCTGGCCGCAGCCAACCCGGCACTG of aa 1083-1116 in pGRN125. Oligo
CCCTCAGAC creates a NheI site.
RT3A ACTCAGGCCCGGCCCCCGCCACACG 60 Mutagenesis oligo to create a deletion
CTAGCGAGACCAAGCACTTCCTCTAC of aa 192-323 in pGRN125. Oligo
TCCTCAGGC creates a NheI site.
RT4A ACTCAGGCCCGGCCCCCGCCACACG 60 Mutagenesis oligo to create a deletion
CTAGCGTGGTGTCACCTGCCAGACCC of aa 192-271 in pGRN125. Oligo
GCCGAAGAA creates a NheI site.
RT6A ATCCCCAAGCCTGACGGGCTGCGGC 69 Mutagenesis oligo to create a deletion
CGATTGTTAACATGCTGTTCAGCGTG of aa 638-660 in pGRN125. Oligo
CTCAACTACGAGCGGGCG creates a Hpa I site.
RT8A ACGTACTGCGTGCGTCGGTATGCCGT 63 Mutagenesis oligo to create a deletion
GGTCACAGATCTCCAGCCGTACATGC of aa 748-766 in pGRN125. Oligo
GACAGTTCGTG creates a BgI II site.
RT3A/5 ACTCAGGCCCGGCCCCCGCCACACG 60 Mutagenesis oligo to create a deletion
CTAGCCTGCTCCGCCAGCACAGCAG of aa 192-450 in pGRN125. Oligo
CCCCTGGCAG creates a NheI site.
LM121- GTTCAGATGCCGGCCCACGGCCTATT 63 Mutagenesis oligo to delete aa 930-934.
WG CCCTCTAGATACCCGGACCCTGGAGG Oligo introduces a new XbaI site
TGCAGAGCGAC
LM122- CCCTGGGCCTGCCAGCCCCGGGTGC 50 Mutagenesis oligo to delete aa 222-240.
Nuc CGGCGCTGCCCCTGAGCCGGAGCGG Oligo introduces a new Nae I site
RT3 GCTAGTGGACCCCGAAGGCGTCTGG 60 Mutagenesis oligo to create a deletion
GATGCGAGACCAAGCACTTCCTCTAC of aa2OO-323 in pGRN125
TCCTCAGGC
RT4 GCTAGTGGACCCCGAAGGCGTCTGG 60 Mutagenesis oligo to create a deletion
GATGCGTGGTGTCACCTGCCAGACCC of aa 200-271 in pGRN125
GCCGAAGAA
RT6 GACGGGCTGCGGCCGATTGTGAACA 60 Mutagenesis oligo to create a deletion
TGGACCTGTTCAGCGTGCTCAACTAC of aa 638-660 in pGRN125
GAGCGGGCG
RT8 ACGTACTGCGTGCGTCGGTATGCCGT 60 Mutagenesis oligo to create a deletion
GGTCACCTTGACAGACCTCCAGCCGT of aa 748-764 in pGRN125
ACATGCGA
V. Definitions
The following terms are defined infra to provide additional guidance to one of skill in the practice of the invention:
As used herein, a polypeptide region in a first polypeptide “corresponds” to a region in a second polypeptide when the amino acid sequences of the two regions, or flanking the two regions, are the same or substantially identical. Sequences can be aligned by inspection (e.g., alignment of identical sequences) or by computer implemented alignment of the two sequences. Thus, for example, the residues 192 to 323 of the hTRT polypeptide having the sequence set forth in FIG. 1 “correspond” to residues in the same position in a hTRT polypeptide that differs from the FIG. 1 sequence due to polymorphic variation, or other mutations or deletions (e.g., when the two polypeptides are optimally aligned). Alignments may also be carried out using the GAP computer program, version 6.0 (Devereux et al, 1984, Nucl. Acid. Res. 12:387; available from the University of Wisconsin Genetics Computer Group, Madison, Wis.). The GAP program utilizes the alignment method of Needleham and Wunsch, 1970 J. Mol. Biol. 48: 443-453 as revised by Smith and Waterman, 1981, Adv. Appl. Math 2:482. The preferred default parameters for the GAP program include (1) the weighted comparison matrix of Gribskov and Burgess, 1986, Nuc. Acid. Res. 14:6745 as described by Schwartz and Dayhoff, eds., 1979, Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, pp. 353-358 (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps. Alternatively, alignments can be carried out using the BLAST algorithm, which is described in Altschul et al., 1990, J. Mol. Biol. 215:403-410 using as defaults a wordlength (W) of 11, the BLOSUM62 scoring matrix (see Henikoff & Henikoff, 1989, Proc. Natl. Acad. Sci. USA 89:10915); alignments (B) of 50, expectation (E) of 10, M=5, and N=−4. A modification of BLAST, the “Gapped BLAST” allows gaps to be introduced into the alignments that are returned (Altschul et al., 1997, Nucleic Acids Res 1:3389-3402). Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/).
When referring to an “activity” of an hTRT variant, a variant is considered to be active in an assay of it displays at least 40% of the activity characteristic of the hTRT polypeptide having the sequence set forth in FIG. 1 (“wild type”). A variant is considered to lack activity when it has less that 1% of the “wild type” activity. A variant with greater than 1% activity and less than 40% activity has “intermediate activity.”
As used herein,“conservative substitution,” refers to substitution of amino acids with other amino acids having similar properties (e.g. acidic, basic, positively or negatively charged, polar or non-polar). The following six groups each contain amino acids that are conservative substitutions for one another: 1) alanine (A), serine (S), threonine (T); 2) aspartic acid (D), glutamic acid (E); 3) asparagine (N), glutamine (Q); 4) arginine (R), lysine (K); 5) isoleucine (I), leucine (L), methionine (M), valine (V); and 6) phenylalanine (F), tyrosine (Y), tryptophan (W) (see also, Creighton, 1984, Proteins, W. H. Freeman and Company).
All publications and patent documents cited in this application are incorporated by reference in their entirety and for all purposes to the same extent as if each individual publication or patent document were so individually denoted.
21 1 4015 DNA Homo sapiens CDS (56)..(3454) human telomerase reverse transcriptase (hTRT) cDNA 1 gcagcgctgc gtcctgctgc gcacgtggga agccctggcc ccggccaccc ccgcg atg 58 Met 1 ccg cgc gct ccc cgc tgc cga gcc gtg cgc tcc ctg ctg cgc agc cac 106 Pro Arg Ala Pro Arg Cys Arg Ala Val Arg Ser Leu Leu Arg Ser His 5 10 15 tac cgc gag gtg ctg ccg ctg gcc acg ttc gtg cgg cgc ctg ggg ccc 154 Tyr Arg Glu Val Leu Pro Leu Ala Thr Phe Val Arg Arg Leu Gly Pro 20 25 30 cag ggc tgg cgg ctg gtg cag cgc ggg gac ccg gcg gct ttc cgc gcg 202 Gln Gly Trp Arg Leu Val Gln Arg Gly Asp Pro Ala Ala Phe Arg Ala 35 40 45 ctg gtg gcc cag tgc ctg gtg tgc gtg ccc tgg gac gca cgg ccg ccc 250 Leu Val Ala Gln Cys Leu Val Cys Val Pro Trp Asp Ala Arg Pro Pro 50 55 60 65 ccc gcc gcc ccc tcc ttc cgc cag gtg tcc tgc ctg aag gag ctg gtg 298 Pro Ala Ala Pro Ser Phe Arg Gln Val Ser Cys Leu Lys Glu Leu Val 70 75 80 gcc cga gtg ctg cag agg ctg tgc gag cgc ggc gcg aag aac gtg ctg 346 Ala Arg Val Leu Gln Arg Leu Cys Glu Arg Gly Ala Lys Asn Val Leu 85 90 95 gcc ttc ggc ttc gcg ctg ctg gac ggg gcc cgc ggg ggc ccc ccc gag 394 Ala Phe Gly Phe Ala Leu Leu Asp Gly Ala Arg Gly Gly Pro Pro Glu 100 105 110 gcc ttc acc acc agc gtg cgc agc tac ctg ccc aac acg gtg acc gac 442 Ala Phe Thr Thr Ser Val Arg Ser Tyr Leu Pro Asn Thr Val Thr Asp 115 120 125 gca ctg cgg ggg agc ggg gcg tgg ggg ctg ctg ctg cgc cgc gtg ggc 490 Ala Leu Arg Gly Ser Gly Ala Trp Gly Leu Leu Leu Arg Arg Val Gly 130 135 140 145 gac gac gtg ctg gtt cac ctg ctg gca cgc tgc gcg ctc ttt gtg ctg 538 Asp Asp Val Leu Val His Leu Leu Ala Arg Cys Ala Leu Phe Val Leu 150 155 160 gtg gct ccc agc tgc gcc tac cag gtg tgc ggg ccg ccg ctg tac cag 586 Val Ala Pro Ser Cys Ala Tyr Gln Val Cys Gly Pro Pro Leu Tyr Gln 165 170 175 ctc ggc gct gcc act cag gcc cgg ccc ccg cca cac gct agt gga ccc 634 Leu Gly Ala Ala Thr Gln Ala Arg Pro Pro Pro His Ala Ser Gly Pro 180 185 190 cga agg cgt ctg gga tgc gaa cgg gcc tgg aac cat agc gtc agg gag 682 Arg Arg Arg Leu Gly Cys Glu Arg Ala Trp Asn His Ser Val Arg Glu 195 200 205 gcc ggg gtc ccc ctg ggc ctg cca gcc ccg ggt gcg agg agg cgc ggg 730 Ala Gly Val Pro Leu Gly Leu Pro Ala Pro Gly Ala Arg Arg Arg Gly 210 215 220 225 ggc agt gcc agc cga agt ctg ccg ttg ccc aag agg ccc agg cgt ggc 778 Gly Ser Ala Ser Arg Ser Leu Pro Leu Pro Lys Arg Pro Arg Arg Gly 230 235 240 gct gcc cct gag ccg gag cgg acg ccc gtt ggg cag ggg tcc tgg gcc 826 Ala Ala Pro Glu Pro Glu Arg Thr Pro Val Gly Gln Gly Ser Trp Ala 245 250 255 cac ccg ggc agg acg cgt gga ccg agt gac cgt ggt ttc tgt gtg gtg 874 His Pro Gly Arg Thr Arg Gly Pro Ser Asp Arg Gly Phe Cys Val Val 260 265 270 tca cct gcc aga ccc gcc gaa gaa gcc acc tct ttg gag ggt gcg ctc 922 Ser Pro Ala Arg Pro Ala Glu Glu Ala Thr Ser Leu Glu Gly Ala Leu 275 280 285 tct ggc acg cgc cac tcc cac cca tcc gtg ggc cgc cag cac cac gcg 970 Ser Gly Thr Arg His Ser His Pro Ser Val Gly Arg Gln His His Ala 290 295 300 305 ggc ccc cca tcc aca tcg cgg cca cca cgt ccc tgg gac acg cct tgt 1018 Gly Pro Pro Ser Thr Ser Arg Pro Pro Arg Pro Trp Asp Thr Pro Cys 310 315 320 ccc ccg gtg tac gcc gag acc aag cac ttc ctc tac tcc tca ggc gac 1066 Pro Pro Val Tyr Ala Glu Thr Lys His Phe Leu Tyr Ser Ser Gly Asp 325 330 335 aag gag cag ctg cgg ccc tcc ttc cta ctc agc tct ctg agg ccc agc 1114 Lys Glu Gln Leu Arg Pro Ser Phe Leu Leu Ser Ser Leu Arg Pro Ser 340 345 350 ctg act ggc gct cgg agg ctc gtg gag acc atc ttt ctg ggt tcc agg 1162 Leu Thr Gly Ala Arg Arg Leu Val Glu Thr Ile Phe Leu Gly Ser Arg 355 360 365 ccc tgg atg cca ggg act ccc cgc agg ttg ccc cgc ctg ccc cag cgc 1210 Pro Trp Met Pro Gly Thr Pro Arg Arg Leu Pro Arg Leu Pro Gln Arg 370 375 380 385 tac tgg caa atg cgg ccc ctg ttt ctg gag ctg ctt ggg aac cac gcg 1258 Tyr Trp Gln Met Arg Pro Leu Phe Leu Glu Leu Leu Gly Asn His Ala 390 395 400 cag tgc ccc tac ggg gtg ctc ctc aag acg cac tgc ccg ctg cga gct 1306 Gln Cys Pro Tyr Gly Val Leu Leu Lys Thr His Cys Pro Leu Arg Ala 405 410 415 gcg gtc acc cca gca gcc ggt gtc tgt gcc cgg gag aag ccc cag ggc 1354 Ala Val Thr Pro Ala Ala Gly Val Cys Ala Arg Glu Lys Pro Gln Gly 420 425 430 tct gtg gcg gcc ccc gag gag gag gac aca gac ccc cgt cgc ctg gtg 1402 Ser Val Ala Ala Pro Glu Glu Glu Asp Thr Asp Pro Arg Arg Leu Val 435 440 445 cag ctg ctc cgc cag cac agc agc ccc tgg cag gtg tac ggc ttc gtg 1450 Gln Leu Leu Arg Gln His Ser Ser Pro Trp Gln Val Tyr Gly Phe Val 450 455 460 465 cgg gcc tgc ctg cgc cgg ctg gtg ccc cca ggc ctc tgg ggc tcc agg 1498 Arg Ala Cys Leu Arg Arg Leu Val Pro Pro Gly Leu Trp Gly Ser Arg 470 475 480 cac aac gaa cgc cgc ttc ctc agg aac acc aag aag ttc atc tcc ctg 1546 His Asn Glu Arg Arg Phe Leu Arg Asn Thr Lys Lys Phe Ile Ser Leu 485 490 495 ggg aag cat gcc aag ctc tcg ctg cag gag ctg acg tgg aag atg agc 1594 Gly Lys His Ala Lys Leu Ser Leu Gln Glu Leu Thr Trp Lys Met Ser 500 505 510 gtg cgg gac tgc gct tgg ctg cgc agg agc cca ggg gtt ggc tgt gtt 1642 Val Arg Asp Cys Ala Trp Leu Arg Arg Ser Pro Gly Val Gly Cys Val 515 520 525 ccg gcc gca gag cac cgt ctg cgt gag gag atc ctg gcc aag ttc ctg 1690 Pro Ala Ala Glu His Arg Leu Arg Glu Glu Ile Leu Ala Lys Phe Leu 530 535 540 545 cac tgg ctg atg agt gtg tac gtc gtc gag ctg ctc agg tct ttc ttt 1738 His Trp Leu Met Ser Val Tyr Val Val Glu Leu Leu Arg Ser Phe Phe 550 555 560 tat gtc acg gag acc acg ttt caa aag aac agg ctc ttt ttc tac cgg 1786 Tyr Val Thr Glu Thr Thr Phe Gln Lys Asn Arg Leu Phe Phe Tyr Arg 565 570 575 aag agt gtc tgg agc aag ttg caa agc att gga atc aga cag cac ttg 1834 Lys Ser Val Trp Ser Lys Leu Gln Ser Ile Gly Ile Arg Gln His Leu 580 585 590 aag agg gtg cag ctg cgg gag ctg tcg gaa gca gag gtc agg cag cat 1882 Lys Arg Val Gln Leu Arg Glu Leu Ser Glu Ala Glu Val Arg Gln His 595 600 605 cgg gaa gcc agg ccc gcc ctg ctg acg tcc aga ctc cgc ttc atc ccc 1930 Arg Glu Ala Arg Pro Ala Leu Leu Thr Ser Arg Leu Arg Phe Ile Pro 610 615 620 625 aag cct gac ggg ctg cgg ccg att gtg aac atg gac tac gtc gtg gga 1978 Lys Pro Asp Gly Leu Arg Pro Ile Val Asn Met Asp Tyr Val Val Gly 630 635 640 gcc aga acg ttc cgc aga gaa aag agg gcc gag cgt ctc acc tcg agg 2026 Ala Arg Thr Phe Arg Arg Glu Lys Arg Ala Glu Arg Leu Thr Ser Arg 645 650 655 gtg aag gca ctg ttc agc gtg ctc aac tac gag cgg gcg cgg cgc ccc 2074 Val Lys Ala Leu Phe Ser Val Leu Asn Tyr Glu Arg Ala Arg Arg Pro 660 665 670 ggc ctc ctg ggc gcc tct gtg ctg ggc ctg gac gat atc cac agg gcc 2122 Gly Leu Leu Gly Ala Ser Val Leu Gly Leu Asp Asp Ile His Arg Ala 675 680 685 tgg cgc acc ttc gtg ctg cgt gtg cgg gcc cag gac ccg ccg cct gag 2170 Trp Arg Thr Phe Val Leu Arg Val Arg Ala Gln Asp Pro Pro Pro Glu 690 695 700 705 ctg tac ttt gtc aag gtg gat gtg acg ggc gcg tac gac acc atc ccc 2218 Leu Tyr Phe Val Lys Val Asp Val Thr Gly Ala Tyr Asp Thr Ile Pro 710 715 720 cag gac agg ctc acg gag gtc atc gcc agc atc atc aaa ccc cag aac 2266 Gln Asp Arg Leu Thr Glu Val Ile Ala Ser Ile Ile Lys Pro Gln Asn 725 730 735 acg tac tgc gtg cgt cgg tat gcc gtg gtc cag aag gcc gcc cat ggg 2314 Thr Tyr Cys Val Arg Arg Tyr Ala Val Val Gln Lys Ala Ala His Gly 740 745 750 cac gtc cgc aag gcc ttc aag agc cac gtc tct acc ttg aca gac ctc 2362 His Val Arg Lys Ala Phe Lys Ser His Val Ser Thr Leu Thr Asp Leu 755 760 765 cag ccg tac atg cga cag ttc gtg gct cac ctg cag gag acc agc ccg 2410 Gln Pro Tyr Met Arg Gln Phe Val Ala His Leu Gln Glu Thr Ser Pro 770 775 780 785 ctg agg gat gcc gtc gtc atc gag cag agc tcc tcc ctg aat gag gcc 2458 Leu Arg Asp Ala Val Val Ile Glu Gln Ser Ser Ser Leu Asn Glu Ala 790 795 800 agc agt ggc ctc ttc gac gtc ttc cta cgc ttc atg tgc cac cac gcc 2506 Ser Ser Gly Leu Phe Asp Val Phe Leu Arg Phe Met Cys His His Ala 805 810 815 gtg cgc atc agg ggc aag tcc tac gtc cag tgc cag ggg atc ccg cag 2554 Val Arg Ile Arg Gly Lys Ser Tyr Val Gln Cys Gln Gly Ile Pro Gln 820 825 830 ggc tcc atc ctc tcc acg ctg ctc tgc agc ctg tgc tac ggc gac atg 2602 Gly Ser Ile Leu Ser Thr Leu Leu Cys Ser Leu Cys Tyr Gly Asp Met 835 840 845 gag aac aag ctg ttt gcg ggg att cgg cgg gac ggg ctg ctc ctg cgt 2650 Glu Asn Lys Leu Phe Ala Gly Ile Arg Arg Asp Gly Leu Leu Leu Arg 850 855 860 865 ttg gtg gat gat ttc ttg ttg gtg aca cct cac ctc acc cac gcg aaa 2698 Leu Val Asp Asp Phe Leu Leu Val Thr Pro His Leu Thr His Ala Lys 870 875 880 acc ttc ctc agg acc ctg gtc cga ggt gtc cct gag tat ggc tgc gtg 2746 Thr Phe Leu Arg Thr Leu Val Arg Gly Val Pro Glu Tyr Gly Cys Val 885 890 895 gtg aac ttg cgg aag aca gtg gtg aac ttc cct gta gaa gac gag gcc 2794 Val Asn Leu Arg Lys Thr Val Val Asn Phe Pro Val Glu Asp Glu Ala 900 905 910 ctg ggt ggc acg gct ttt gtt cag atg ccg gcc cac ggc cta ttc ccc 2842 Leu Gly Gly Thr Ala Phe Val Gln Met Pro Ala His Gly Leu Phe Pro 915 920 925 tgg tgc ggc ctg ctg ctg gat acc cgg acc ctg gag gtg cag agc gac 2890 Trp Cys Gly Leu Leu Leu Asp Thr Arg Thr Leu Glu Val Gln Ser Asp 930 935 940 945 tac tcc agc tat gcc cgg acc tcc atc aga gcc agt ctc acc ttc aac 2938 Tyr Ser Ser Tyr Ala Arg Thr Ser Ile Arg Ala Ser Leu Thr Phe Asn 950 955 960 cgc ggc ttc aag gct ggg agg aac atg cgt cgc aaa ctc ttt ggg gtc 2986 Arg Gly Phe Lys Ala Gly Arg Asn Met Arg Arg Lys Leu Phe Gly Val 965 970 975 ttg cgg ctg aag tgt cac agc ctg ttt ctg gat ttg cag gtg aac agc 3034 Leu Arg Leu Lys Cys His Ser Leu Phe Leu Asp Leu Gln Val Asn Ser 980 985 990 ctc cag acg gtg tgc acc aac atc tac aag atc ctc ctg ctg cag gcg 3082 Leu Gln Thr Val Cys Thr Asn Ile Tyr Lys Ile Leu Leu Leu Gln Ala 995 1000 1005 tac agg ttt cac gca tgt gtg ctg cag ctc cca ttt cat cag caa gtt 3130 Tyr Arg Phe His Ala Cys Val Leu Gln Leu Pro Phe His Gln Gln Val 1010 1015 1020 1025 tgg aag aac ccc aca ttt ttc ctg cgc gtc atc tct gac acg gcc tcc 3178 Trp Lys Asn Pro Thr Phe Phe Leu Arg Val Ile Ser Asp Thr Ala Ser 1030 1035 1040 ctc tgc tac tcc atc ctg aaa gcc aag aac gca ggg atg tcg ctg ggg 3226 Leu Cys Tyr Ser Ile Leu Lys Ala Lys Asn Ala Gly Met Ser Leu Gly 1045 1050 1055 gcc aag ggc gcc gcc ggc cct ctg ccc tcc gag gcc gtg cag tgg ctg 3274 Ala Lys Gly Ala Ala Gly Pro Leu Pro Ser Glu Ala Val Gln Trp Leu 1060 1065 1070 tgc cac caa gca ttc ctg ctc aag ctg act cga cac cgt gtc acc tac 3322 Cys His Gln Ala Phe Leu Leu Lys Leu Thr Arg His Arg Val Thr Tyr 1075 1080 1085 gtg cca ctc ctg ggg tca ctc agg aca gcc cag acg cag ctg agt cgg 3370 Val Pro Leu Leu Gly Ser Leu Arg Thr Ala Gln Thr Gln Leu Ser Arg 1090 1095 1100 1105 aag ctc ccg ggg acg acg ctg act gcc ctg gag gcc gca gcc aac ccg 3418 Lys Leu Pro Gly Thr Thr Leu Thr Ala Leu Glu Ala Ala Ala Asn Pro 1110 1115 1120 gca ctg ccc tca gac ttc aag acc atc ctg gac tga tggccacccg 3464 Ala Leu Pro Ser Asp Phe Lys Thr Ile Leu Asp 1125 1130 cccacagcca ggccgagagc agacaccagc agccctgtca cgccgggctc tacgtcccag 3524 ggagggaggg gcggcccaca cccaggcccg caccgctggg agtctgaggc ctgagtgagt 3584 gtttggccga ggcctgcatg tccggctgaa ggctgagtgt ccggctgagg cctgagcgag 3644 tgtccagcca agggctgagt gtccagcaca cctgccgtct tcacttcccc acaggctggc 3704 gctcggctcc accccagggc cagcttttcc tcaccaggag cccggcttcc actccccaca 3764 taggaatagt ccatccccag attcgccatt gttcacccct cgccctgccc tcctttgcct 3824 tccaccccca ccatccaggt ggagaccctg agaaggaccc tgggagctct gggaatttgg 3884 agtgaccaaa ggtgtgccct gtacacaggc gaggaccctg cacctggatg ggggtccctg 3944 tgggtcaaat tggggggagg tgctgtggga gtaaaatact gaatatatga gtttttcagt 4004 tttgaaaaaa a 4015 2 1132 PRT Homo sapiens 2 Met Pro Arg Ala Pro Arg Cys Arg Ala Val Arg Ser Leu Leu Arg Ser 1 5 10 15 His Tyr Arg Glu Val Leu Pro Leu Ala Thr Phe Val Arg Arg Leu Gly 20 25 30 Pro Gln Gly Trp Arg Leu Val Gln Arg Gly Asp Pro Ala Ala Phe Arg 35 40 45 Ala Leu Val Ala Gln Cys Leu Val Cys Val Pro Trp Asp Ala Arg Pro 50 55 60 Pro Pro Ala Ala Pro Ser Phe Arg Gln Val Ser Cys Leu Lys Glu Leu 65 70 75 80 Val Ala Arg Val Leu Gln Arg Leu Cys Glu Arg Gly Ala Lys Asn Val 85 90 95 Leu Ala Phe Gly Phe Ala Leu Leu Asp Gly Ala Arg Gly Gly Pro Pro 100 105 110 Glu Ala Phe Thr Thr Ser Val Arg Ser Tyr Leu Pro Asn Thr Val Thr 115 120 125 Asp Ala Leu Arg Gly Ser Gly Ala Trp Gly Leu Leu Leu Arg Arg Val 130 135 140 Gly Asp Asp Val Leu Val His Leu Leu Ala Arg Cys Ala Leu Phe Val 145 150 155 160 Leu Val Ala Pro Ser Cys Ala Tyr Gln Val Cys Gly Pro Pro Leu Tyr 165 170 175 Gln Leu Gly Ala Ala Thr Gln Ala Arg Pro Pro Pro His Ala Ser Gly 180 185 190 Pro Arg Arg Arg Leu Gly Cys Glu Arg Ala Trp Asn His Ser Val Arg 195 200 205 Glu Ala Gly Val Pro Leu Gly Leu Pro Ala Pro Gly Ala Arg Arg Arg 210 215 220 Gly Gly Ser Ala Ser Arg Ser Leu Pro Leu Pro Lys Arg Pro Arg Arg 225 230 235 240 Gly Ala Ala Pro Glu Pro Glu Arg Thr Pro Val Gly Gln Gly Ser Trp 245 250 255 Ala His Pro Gly Arg Thr Arg Gly Pro Ser Asp Arg Gly Phe Cys Val 260 265 270 Val Ser Pro Ala Arg Pro Ala Glu Glu Ala Thr Ser Leu Glu Gly Ala 275 280 285 Leu Ser Gly Thr Arg His Ser His Pro Ser Val Gly Arg Gln His His 290 295 300 Ala Gly Pro Pro Ser Thr Ser Arg Pro Pro Arg Pro Trp Asp Thr Pro 305 310 315 320 Cys Pro Pro Val Tyr Ala Glu Thr Lys His Phe Leu Tyr Ser Ser Gly 325 330 335 Asp Lys Glu Gln Leu Arg Pro Ser Phe Leu Leu Ser Ser Leu Arg Pro 340 345 350 Ser Leu Thr Gly Ala Arg Arg Leu Val Glu Thr Ile Phe Leu Gly Ser 355 360 365 Arg Pro Trp Met Pro Gly Thr Pro Arg Arg Leu Pro Arg Leu Pro Gln 370 375 380 Arg Tyr Trp Gln Met Arg Pro Leu Phe Leu Glu Leu Leu Gly Asn His 385 390 395 400 Ala Gln Cys Pro Tyr Gly Val Leu Leu Lys Thr His Cys Pro Leu Arg 405 410 415 Ala Ala Val Thr Pro Ala Ala Gly Val Cys Ala Arg Glu Lys Pro Gln 420 425 430 Gly Ser Val Ala Ala Pro Glu Glu Glu Asp Thr Asp Pro Arg Arg Leu 435 440 445 Val Gln Leu Leu Arg Gln His Ser Ser Pro Trp Gln Val Tyr Gly Phe 450 455 460 Val Arg Ala Cys Leu Arg Arg Leu Val Pro Pro Gly Leu Trp Gly Ser 465 470 475 480 Arg His Asn Glu Arg Arg Phe Leu Arg Asn Thr Lys Lys Phe Ile Ser 485 490 495 Leu Gly Lys His Ala Lys Leu Ser Leu Gln Glu Leu Thr Trp Lys Met 500 505 510 Ser Val Arg Asp Cys Ala Trp Leu Arg Arg Ser Pro Gly Val Gly Cys 515 520 525 Val Pro Ala Ala Glu His Arg Leu Arg Glu Glu Ile Leu Ala Lys Phe 530 535 540 Leu His Trp Leu Met Ser Val Tyr Val Val Glu Leu Leu Arg Ser Phe 545 550 555 560 Phe Tyr Val Thr Glu Thr Thr Phe Gln Lys Asn Arg Leu Phe Phe Tyr 565 570 575 Arg Lys Ser Val Trp Ser Lys Leu Gln Ser Ile Gly Ile Arg Gln His 580 585 590 Leu Lys Arg Val Gln Leu Arg Glu Leu Ser Glu Ala Glu Val Arg Gln 595 600 605 His Arg Glu Ala Arg Pro Ala Leu Leu Thr Ser Arg Leu Arg Phe Ile 610 615 620 Pro Lys Pro Asp Gly Leu Arg Pro Ile Val Asn Met Asp Tyr Val Val 625 630 635 640 Gly Ala Arg Thr Phe Arg Arg Glu Lys Arg Ala Glu Arg Leu Thr Ser 645 650 655 Arg Val Lys Ala Leu Phe Ser Val Leu Asn Tyr Glu Arg Ala Arg Arg 660 665 670 Pro Gly Leu Leu Gly Ala Ser Val Leu Gly Leu Asp Asp Ile His Arg 675 680 685 Ala Trp Arg Thr Phe Val Leu Arg Val Arg Ala Gln Asp Pro Pro Pro 690 695 700 Glu Leu Tyr Phe Val Lys Val Asp Val Thr Gly Ala Tyr Asp Thr Ile 705 710 715 720 Pro Gln Asp Arg Leu Thr Glu Val Ile Ala Ser Ile Ile Lys Pro Gln 725 730 735 Asn Thr Tyr Cys Val Arg Arg Tyr Ala Val Val Gln Lys Ala Ala His 740 745 750 Gly His Val Arg Lys Ala Phe Lys Ser His Val Ser Thr Leu Thr Asp 755 760 765 Leu Gln Pro Tyr Met Arg Gln Phe Val Ala His Leu Gln Glu Thr Ser 770 775 780 Pro Leu Arg Asp Ala Val Val Ile Glu Gln Ser Ser Ser Leu Asn Glu 785 790 795 800 Ala Ser Ser Gly Leu Phe Asp Val Phe Leu Arg Phe Met Cys His His 805 810 815 Ala Val Arg Ile Arg Gly Lys Ser Tyr Val Gln Cys Gln Gly Ile Pro 820 825 830 Gln Gly Ser Ile Leu Ser Thr Leu Leu Cys Ser Leu Cys Tyr Gly Asp 835 840 845 Met Glu Asn Lys Leu Phe Ala Gly Ile Arg Arg Asp Gly Leu Leu Leu 850 855 860 Arg Leu Val Asp Asp Phe Leu Leu Val Thr Pro His Leu Thr His Ala 865 870 875 880 Lys Thr Phe Leu Arg Thr Leu Val Arg Gly Val Pro Glu Tyr Gly Cys 885 890 895 Val Val Asn Leu Arg Lys Thr Val Val Asn Phe Pro Val Glu Asp Glu 900 905 910 Ala Leu Gly Gly Thr Ala Phe Val Gln Met Pro Ala His Gly Leu Phe 915 920 925 Pro Trp Cys Gly Leu Leu Leu Asp Thr Arg Thr Leu Glu Val Gln Ser 930 935 940 Asp Tyr Ser Ser Tyr Ala Arg Thr Ser Ile Arg Ala Ser Leu Thr Phe 945 950 955 960 Asn Arg Gly Phe Lys Ala Gly Arg Asn Met Arg Arg Lys Leu Phe Gly 965 970 975 Val Leu Arg Leu Lys Cys His Ser Leu Phe Leu Asp Leu Gln Val Asn 980 985 990 Ser Leu Gln Thr Val Cys Thr Asn Ile Tyr Lys Ile Leu Leu Leu Gln 995 1000 1005 Ala Tyr Arg Phe His Ala Cys Val Leu Gln Leu Pro Phe His Gln Gln 1010 1015 1020 Val Trp Lys Asn Pro Thr Phe Phe Leu Arg Val Ile Ser Asp Thr Ala 1025 1030 1035 1040 Ser Leu Cys Tyr Ser Ile Leu Lys Ala Lys Asn Ala Gly Met Ser Leu 1045 1050 1055 Gly Ala Lys Gly Ala Ala Gly Pro Leu Pro Ser Glu Ala Val Gln Trp 1060 1065 1070 Leu Cys His Gln Ala Phe Leu Leu Lys Leu Thr Arg His Arg Val Thr 1075 1080 1085 Tyr Val Pro Leu Leu Gly Ser Leu Arg Thr Ala Gln Thr Gln Leu Ser 1090 1095 1100 Arg Lys Leu Pro Gly Thr Thr Leu Thr Ala Leu Glu Ala Ala Ala Asn 1105 1110 1115 1120 Pro Ala Leu Pro Ser Asp Phe Lys Thr Ile Leu Asp 1125 1130 3 6 PRT Homo sapiens PEPTIDE (1)..(6) amino acid positions 560-565 from hTRT 3 Phe Phe Tyr Val Thr Glu 1 5 4 14 PRT Homo sapiens PEPTIDE (1)..(14) amino acid positions 405-418 from hTRT 4 Tyr Gly Val Leu Leu Lys Thr His Cys Pro Leu Arg Ala Ala 1 5 10 5 4 PRT Homo sapiens PEPTIDE (1)..(4) amino acid positions 561-564 from hTRT 5 Phe Tyr Val Thr 1 6 25 DNA Artificial Sequence Description of Artificial SequenceRT1 oligo 6 gaaggccgcc cacgggcacg tccgc 25 7 32 DNA Artificial Sequence Description of Artificial SequenceRT2 oligo 7 cccggccacc ccagccatgg cgcgcgctcc cc 32 8 60 DNA Artificial Sequence Description of Artificial SequenceRT5 oligo 8 tacggggtgc tcctcaagac gcactgcccg ctgctccgcc agcacagcag cccctggcag 60 9 60 DNA Artificial Sequence Description of Artificial SequenceRT10 oligo 9 tactccatcc tgaaagccaa gaacgcaggg ctgtgccacc aagcattcct gctcaagctg 60 10 60 DNA Artificial Sequence Description of Artificial SequenceRT11 oligo 10 ctgtgccacc aagcattcct gctcaagctg gccgcagcca acccggcact gccctcagac 60 11 60 DNA Artificial Sequence Description of Artificial SequenceRT3A oligo 11 actcaggccc ggcccccgcc acacgctagc gagaccaagc acttcctcta ctcctcaggc 60 12 60 DNA Artificial Sequence Description of Artificial SequenceRT4A oligo 12 actcaggccc ggcccccgcc acacgctagc gtggtgtcac ctgccagacc cgccgaagaa 60 13 69 DNA Artificial Sequence Description of Artificial SequenceRT6A oligo 13 atccccaagc ctgacgggct gcggccgatt gttaacatgc tgttcagcgt gctcaactac 60 gagcgggcg 69 14 63 DNA Artificial Sequence Description of Artificial SequenceRT8A oligo 14 acgtactgcg tgcgtcggta tgccgtggtc acagatctcc agccgtacat gcgacagttc 60 gtg 63 15 60 DNA Artificial Sequence Description of Artificial SequenceRT3A/5 oligo 15 actcaggccc ggcccccgcc acacgctagc ctgctccgcc agcacagcag cccctggcag 60 16 63 DNA Artificial Sequence Description of Artificial SequenceLM121-WG oligo 16 gttcagatgc cggcccacgg cctattccct ctagataccc ggaccctgga ggtgcagagc 60 gac 63 17 50 DNA Artificial Sequence Description of Artificial SequenceLM122-Nuc oligo 17 ccctgggcct gccagccccg ggtgccggcg ctgcccctga gccggagcgg 50 18 60 DNA Artificial Sequence Description of Artificial SequenceRT3 oligo 18 gctagtggac cccgaaggcg tctgggatgc gagaccaagc acttcctcta ctcctcaggc 60 19 60 DNA Artificial Sequence Description of Artificial SequenceRT4 oligo 19 gctagtggac cccgaaggcg tctgggatgc gtggtgtcac ctgccagacc cgccgaagaa 60 20 60 DNA Artificial Sequence Description of Artificial SequenceRT6 oligo 20 gacgggctgc ggccgattgt gaacatggac ctgttcagcg tgctcaacta cgagcgggcg 60 21 60 DNA Artificial Sequence Description of Artificial SequenceRT8 oligo 21 acgtactgcg tgcgtcggta tgccgtggtc accttgacag acctccagcc gtacatgcga 60

Claims (11)

What is claimed is:
1. A polynucleotide encoding a variant of human telomerase reverse transcriptase (hTRT), said variant having processive catalytic activity and comprising a deletion of at least 10 amino acids from region 192-323 or 415-450 of SEQ. ID NO:2.
2. The polynucleotide of claim 1, wherein the variant comprises a deletion of at least 25 amino acids from region 192-323 or 415-450 of SEQ. ID NO:2.
3. The polynucleotide of claim 1, further comprising a promoter sequence operably linked to the nucleotide sequence encoding the hTRT variant.
4. The polynucleotide of claim 1 that has a deletion of at least one region encoding exactly amino acids 192-323, 200-323, 200-271, 222-240, or 415-450 of SEQ. ID NO:2.
5. The polynucleotide of claim 1 that does not comprise a deletion in the region encoding amino acids 415-450.
6. The polynucleotide of claim 5, further comprising a promoter sequence operably linked to the nucleotide sequence encoding the hTRT variant.
7. A method for increasing the proliferative capacity of a human cell in vitro, comprising expressing the polynucleotide of claim 6 in the cell, thereby increasing its proliferative capacity.
8. A method for increasing the proliferative capacity of a human cell in vitro, comprising expressing the polynucleotide of claim 3 in the cell, thereby increasing its proliferative capacity.
9. A method for producing a variant telomerase reverse transcriptase, comprising expressing the polynucleotide of claim 1 in a host cell or in a cell-free expression system.
10. A cell comprising the polynucleotide of claim 1.
11. The cell of claim 10, that is a human cell.
US09/128,354 1998-03-31 1998-08-03 Human telomerase catalytic subunit variants Expired - Lifetime US6337200B1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US09/128,354 US6337200B1 (en) 1998-03-31 1998-08-03 Human telomerase catalytic subunit variants
PCT/US1999/007097 WO1999050386A2 (en) 1998-03-31 1999-03-31 Human telomerase catayltic subunit variants
AU33752/99A AU3375299A (en) 1998-03-31 1999-03-31 Human telomerase catayltic subunit variants
US09/990,080 US7091021B2 (en) 1998-03-31 2001-11-21 Inactive variants of the human telomerase catalytic subunit
US11/504,402 US8796438B2 (en) 1998-03-31 2006-08-14 Nucleic acids encoding inactive variants of human telomerase
US14/317,418 US20150087064A1 (en) 1998-03-31 2014-06-27 Nucleic acids encoding inactive variants of human telomerase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5286498A 1998-03-31 1998-03-31
US09/128,354 US6337200B1 (en) 1998-03-31 1998-08-03 Human telomerase catalytic subunit variants

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US5286498A Continuation-In-Part 1998-03-31 1998-03-31

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US09/990,080 Continuation US7091021B2 (en) 1998-03-31 2001-11-21 Inactive variants of the human telomerase catalytic subunit

Publications (1)

Publication Number Publication Date
US6337200B1 true US6337200B1 (en) 2002-01-08

Family

ID=26731185

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/128,354 Expired - Lifetime US6337200B1 (en) 1998-03-31 1998-08-03 Human telomerase catalytic subunit variants
US09/990,080 Expired - Lifetime US7091021B2 (en) 1998-03-31 2001-11-21 Inactive variants of the human telomerase catalytic subunit
US11/504,402 Expired - Fee Related US8796438B2 (en) 1998-03-31 2006-08-14 Nucleic acids encoding inactive variants of human telomerase
US14/317,418 Abandoned US20150087064A1 (en) 1998-03-31 2014-06-27 Nucleic acids encoding inactive variants of human telomerase

Family Applications After (3)

Application Number Title Priority Date Filing Date
US09/990,080 Expired - Lifetime US7091021B2 (en) 1998-03-31 2001-11-21 Inactive variants of the human telomerase catalytic subunit
US11/504,402 Expired - Fee Related US8796438B2 (en) 1998-03-31 2006-08-14 Nucleic acids encoding inactive variants of human telomerase
US14/317,418 Abandoned US20150087064A1 (en) 1998-03-31 2014-06-27 Nucleic acids encoding inactive variants of human telomerase

Country Status (3)

Country Link
US (4) US6337200B1 (en)
AU (1) AU3375299A (en)
WO (1) WO1999050386A2 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020164786A1 (en) * 1996-10-01 2002-11-07 Cech Thomas R. Novel telomerase
US20020187471A1 (en) * 1996-10-01 2002-12-12 Cech Thomas R. Novel telomerase
US20030009019A1 (en) * 1996-10-01 2003-01-09 Thomas R. Cech Novel telomerase
US20030096344A1 (en) * 1996-10-01 2003-05-22 Cech Thomas R. Human telomerase catalytic subunit: diagnostic and therapeutic methods
US6767719B1 (en) 1997-11-26 2004-07-27 Geron Corporation Mouse telomerase reverse transcriptase
US20040242529A1 (en) * 1997-04-18 2004-12-02 Geron Corporation Vector encoding inactivated telomerase for treating cancer
US20040248246A1 (en) * 2003-06-05 2004-12-09 The Government Of The United States Compositions and methods for enhancing differential expression
US20040247613A1 (en) * 1997-04-18 2004-12-09 Geron Corporation Treating cancer using a telomerase vaccine
US20050013825A1 (en) * 1997-04-18 2005-01-20 Geron Corporation Vaccine containing the catalytic subunit of telomerase for treating cancer
US20050070492A1 (en) * 1997-04-25 2005-03-31 Cech Thomas R. Treating cancer using antisense oligonucleotides against telomerase reverse transcriptase
US20060040307A1 (en) * 1997-04-18 2006-02-23 Geron Corporation Human telomerase catalytic subunit
US20060052324A1 (en) * 2004-08-05 2006-03-09 Artandi Steven E Methods and compositions for cell activation
US20060275267A1 (en) * 1998-03-31 2006-12-07 Morin Gregg B Nucleic acids encoding inactive variants of human telomerase
US20070020722A1 (en) * 2003-04-30 2007-01-25 Counter Christopher M Telomere elongation
US7390891B1 (en) * 1996-11-15 2008-06-24 Amgen Inc. Polynucleotides encoding a telomerase component TP2
US7517971B1 (en) * 1996-10-01 2009-04-14 Geron Corporation Muteins of human telomerase reverse transcriptase lacking telomerase catalytic activity
US7879609B2 (en) 1996-10-01 2011-02-01 Geron Corporation Regulatory segments of the human gene for telomerase reverse transcriptase
US10251944B2 (en) 2016-01-19 2019-04-09 Pfizer Inc. Cancer vaccines
US10383928B2 (en) 2010-02-16 2019-08-20 Ultimovacs As Telomerase polypeptide vaccine for treating cancer

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7030211B1 (en) 1998-07-08 2006-04-18 Gemvax As Antigenic peptides derived from telomerase
GB0031430D0 (en) * 2000-12-22 2001-02-07 Norsk Hydro As Polypeptides
US7166692B2 (en) 2003-03-04 2007-01-23 Canbrex Bio Science Walkersville, Inc. Intracellular delivery of small molecules, proteins, and nucleic acids
CN102387813B (en) 2009-04-02 2018-07-31 瓦克松生物技术公司 Identification, optimization and the purposes of hidden HLA-A24 epitopes for immunization therapy
CN109069575A (en) * 2016-02-23 2018-12-21 毛里齐奥·扎内蒂 universal cancer vaccine
KR20210057024A (en) * 2018-09-06 2021-05-20 더 보드 오브 리젠츠 오브 더 유니버시티 오브 텍사스 시스템 Telomerase complete enzyme complex and method of use thereof

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154598B (en) 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4554101A (en) 1981-01-09 1985-11-19 New York Blood Center, Inc. Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5001225A (en) 1986-12-08 1991-03-19 Georgetown University Monoclonal antibodies to a pan-malarial antigen
US4889806A (en) 1987-04-15 1989-12-26 Washington University Large DNA cloning system based on yeast artificial chromosomes
US5075227A (en) 1989-03-07 1991-12-24 Zymogenetics, Inc. Directional cloning
US5489508A (en) 1992-05-13 1996-02-06 University Of Texas System Board Of Regents Therapy and diagnosis of conditions related to telomere length and/or telomerase activity
US5639613A (en) 1992-05-13 1997-06-17 Board Of Regents, University Of Texas System Methods for cancer diagnosis and prognosis
US6120764A (en) 1993-06-24 2000-09-19 Advec, Inc. Adenoviruses for control of gene expression
US5919676A (en) 1993-06-24 1999-07-06 Advec, Inc. Adenoviral vector system comprising Cre-loxP recombination
US6140087A (en) 1993-06-24 2000-10-31 Advec, Inc. Adenovirus vectors for gene therapy
US5583016A (en) 1994-07-07 1996-12-10 Geron Corporation Mammalian telomerase
US5597697A (en) 1994-09-30 1997-01-28 Diamond; Paul Screening assay for inhibitors and activators of RNA and DNA-dependent nucleic acid polymerases
US5698686A (en) 1994-10-20 1997-12-16 Arch Development Corporation Yeast telomerase compositions
US6545133B1 (en) 1995-08-04 2003-04-08 Geron Corporation Methods for purifying telomerase
US6517834B1 (en) 1995-08-04 2003-02-11 Geron Corporation Purified telomerase
US5968506A (en) 1995-08-04 1999-10-19 Geron Corporation Purified telomerase
US5853719A (en) 1996-04-30 1998-12-29 Duke University Methods for treating cancers and pathogen infections using antigen-presenting cells loaded with RNA
US5770422A (en) 1996-07-08 1998-06-23 The Regents Of The University Of California Human telomerase
US5747317A (en) 1996-07-08 1998-05-05 Tularik Inc. Human telomerase RNA interacting protein gene
WO1998007838A1 (en) 1996-08-21 1998-02-26 Mitsubishi Chemical Corporation Higher animal telomerase protein and gene encoding the same
US6808880B2 (en) 1996-10-01 2004-10-26 Geron Corporation Method for detecting polynucleotides encoding telomerase
US7585622B1 (en) * 1996-10-01 2009-09-08 Geron Corporation Increasing the proliferative capacity of cells using telomerase reverse transcriptase
US6610839B1 (en) 1997-08-14 2003-08-26 Geron Corporation Promoter for telomerase reverse transcriptase
DE69723531T3 (en) 1996-10-01 2012-09-06 Geron Corp. Catalytic subunit of human telomerase
US6444650B1 (en) 1996-10-01 2002-09-03 Geron Corporation Antisense compositions for detecting and inhibiting telomerase reverse transcriptase
US6777203B1 (en) 1997-11-19 2004-08-17 Geron Corporation Telomerase promoter driving expression of therapeutic gene sequences
US6093809A (en) 1996-10-01 2000-07-25 University Technology Corporation Telomerase
US6475789B1 (en) 1996-10-01 2002-11-05 University Technology Corporation Human telomerase catalytic subunit: diagnostic and therapeutic methods
US6261836B1 (en) 1996-10-01 2001-07-17 Geron Corporation Telomerase
US6767719B1 (en) 1997-11-26 2004-07-27 Geron Corporation Mouse telomerase reverse transcriptase
US5919656A (en) 1996-11-15 1999-07-06 Amgen Canada Inc. Genes encoding telomerase protein 1
US7390891B1 (en) 1996-11-15 2008-06-24 Amgen Inc. Polynucleotides encoding a telomerase component TP2
WO1998037181A2 (en) * 1997-02-20 1998-08-27 Whitehead Institute For Biomedical Research Telomerase catalytic subunit gene and encoded protein
US20050013825A1 (en) 1997-04-18 2005-01-20 Geron Corporation Vaccine containing the catalytic subunit of telomerase for treating cancer
US7413864B2 (en) 1997-04-18 2008-08-19 Geron Corporation Treating cancer using a telomerase vaccine
US7622549B2 (en) 1997-04-18 2009-11-24 Geron Corporation Human telomerase reverse transcriptase polypeptides
IL133830A0 (en) 1997-07-01 2001-04-30 Cambia Biosystems Llc Vertebrate tolomerase genes and proteins and uses thereof
US7378244B2 (en) 1997-10-01 2008-05-27 Geron Corporation Telomerase promoters sequences for screening telomerase modulators
WO1999027113A1 (en) 1997-11-26 1999-06-03 Geron Corporation Mouse telomerase reverse transcriptase
DE69931600T2 (en) 1998-01-08 2007-05-03 Chugai Seiyaku K.K. NEW GENE WITH A REVERS TRANSCRIPTASE MOTIF
US7402307B2 (en) 1998-03-31 2008-07-22 Geron Corporation Method for identifying and killing cancer cells
US6337200B1 (en) 1998-03-31 2002-01-08 Geron Corporation Human telomerase catalytic subunit variants
EP1068296B1 (en) 1998-03-31 2011-08-10 Geron Corporation Compositions for eliciting an immune response to a telomerase antigen
AU2002342613A1 (en) 2001-05-09 2002-11-25 Geron Corporation Treatment for wounds
US7294708B2 (en) 2002-05-31 2007-11-13 Beijing Institute Of Biotechnology Telomerase reverse transcriptase fragments and uses thereof
US7846904B2 (en) 2003-06-23 2010-12-07 Geron Corporation Compositions and methods for increasing telomerase activity
US20070122501A1 (en) 2003-06-27 2007-05-31 Hong Kong University Of Science And Technology Formulations containing astragalus extracts and uses thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Bodnar et al., "Extension of Life-Span by Introduction of Telomerase into Normal Human Cells" Science 279: 349-352.
Harrington et al., 1997, "Human telomerase contains evolutionarily conserved catalytic and structural subunits"Genes Dev. 11: 3109-3115.
Kilian et al., 1997, "Isolation of a candidate human telomerase catalytic subunit gene, which reveals complex splicing patterns in different cell types" Hum. Mol. Genet. 6: 2011-2019.
Meyerson et al., 1997, "hEST2, the Putative Human Telomerase Catalytic Subunit Gene, Is Up-Regulated in Tumor Cells and during Immortalization" Cell 90: 785-795.
Nakamura et al., 1997, "Telomerase Catalytic Subunit Homologs from Fission Yeast and Human" Science 277: 955.
Weinrich et al., 1997, "Reconstitution of human telomerase with the template RNA component hTR and the catalytic protein subunit hTRT" Nat. Genet. 1997 Dec. 1: 17(4):498-502.

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7517971B1 (en) * 1996-10-01 2009-04-14 Geron Corporation Muteins of human telomerase reverse transcriptase lacking telomerase catalytic activity
US20020164786A1 (en) * 1996-10-01 2002-11-07 Cech Thomas R. Novel telomerase
US20030009019A1 (en) * 1996-10-01 2003-01-09 Thomas R. Cech Novel telomerase
US20030032075A1 (en) * 1996-10-01 2003-02-13 Cech Thomas R. Novel telomerase
US20030044953A1 (en) * 1996-10-01 2003-03-06 Cech Thomas R. Novel telomerase
US20030059787A1 (en) * 1996-10-01 2003-03-27 Cech Thomas R. Novel telomerase
US7005262B2 (en) 1996-10-01 2006-02-28 Geron Corporation Methods for detecting nucleic acids encoding human telomerase reverse transcriptase
US20030100093A1 (en) * 1996-10-01 2003-05-29 Cech Thomas R. Human telomerase catalytic subunit: diagnostic and therapeutic methods
US8222392B2 (en) 1996-10-01 2012-07-17 Geron Corporation Kit for detection of telomerase reverse transcriptase nucleic acids
US6808880B2 (en) 1996-10-01 2004-10-26 Geron Corporation Method for detecting polynucleotides encoding telomerase
US20020187471A1 (en) * 1996-10-01 2002-12-12 Cech Thomas R. Novel telomerase
US7195911B2 (en) 1996-10-01 2007-03-27 Geron Corporation Mammalian cells that have increased proliferative capacity
US7879609B2 (en) 1996-10-01 2011-02-01 Geron Corporation Regulatory segments of the human gene for telomerase reverse transcriptase
US7585622B1 (en) 1996-10-01 2009-09-08 Geron Corporation Increasing the proliferative capacity of cells using telomerase reverse transcriptase
US7560437B2 (en) 1996-10-01 2009-07-14 Geron Corporation Nucleic acid compositions for eliciting an immune response against telomerase reverse transcriptase
US6921664B2 (en) 1996-10-01 2005-07-26 Regents Of The University Of Colorado Telomerase
US6927285B2 (en) 1996-10-01 2005-08-09 Geron Corporation Genes for human telomerase reverse transcriptase and telomerase variants
US7056513B2 (en) 1996-10-01 2006-06-06 Geron Corporation Telomerase
US20030096344A1 (en) * 1996-10-01 2003-05-22 Cech Thomas R. Human telomerase catalytic subunit: diagnostic and therapeutic methods
US20090269739A1 (en) * 1996-10-01 2009-10-29 Geron Corporation Kit for detection of telomerase reverse transcriptase nucleic acids
US7285639B2 (en) 1996-10-01 2007-10-23 Geron Corporation Antibody to telomerase reverse transcriptase
US7390891B1 (en) * 1996-11-15 2008-06-24 Amgen Inc. Polynucleotides encoding a telomerase component TP2
US20040242529A1 (en) * 1997-04-18 2004-12-02 Geron Corporation Vector encoding inactivated telomerase for treating cancer
US7622549B2 (en) 1997-04-18 2009-11-24 Geron Corporation Human telomerase reverse transcriptase polypeptides
US7262288B1 (en) * 1997-04-18 2007-08-28 Geron Corporation Nucleic acids encoding human telomerase reverse transcriptase and related homologs
US8236774B2 (en) 1997-04-18 2012-08-07 Geron Corporation Human telomerase catalytic subunit
US20040247613A1 (en) * 1997-04-18 2004-12-09 Geron Corporation Treating cancer using a telomerase vaccine
US20060040307A1 (en) * 1997-04-18 2006-02-23 Geron Corporation Human telomerase catalytic subunit
US7413864B2 (en) 1997-04-18 2008-08-19 Geron Corporation Treating cancer using a telomerase vaccine
US20080279871A1 (en) * 1997-04-18 2008-11-13 Geron Corporation Immunogenic composition
US8709995B2 (en) 1997-04-18 2014-04-29 Geron Corporation Method for eliciting an immune response to human telomerase reverse transcriptase
US7750121B2 (en) 1997-04-18 2010-07-06 Geron Corporation Antibody to telomerase reverse transcriptive
US20050013825A1 (en) * 1997-04-18 2005-01-20 Geron Corporation Vaccine containing the catalytic subunit of telomerase for treating cancer
US20050070492A1 (en) * 1997-04-25 2005-03-31 Cech Thomas R. Treating cancer using antisense oligonucleotides against telomerase reverse transcriptase
US7297488B2 (en) 1997-04-25 2007-11-20 Geron Corporation Identifying and testing antisense oligonucleotides that inhibit telomerase reverse transcriptase
US6767719B1 (en) 1997-11-26 2004-07-27 Geron Corporation Mouse telomerase reverse transcriptase
US8796438B2 (en) 1998-03-31 2014-08-05 Geron Corporation Nucleic acids encoding inactive variants of human telomerase
US20060275267A1 (en) * 1998-03-31 2006-12-07 Morin Gregg B Nucleic acids encoding inactive variants of human telomerase
US8771982B2 (en) 2003-04-30 2014-07-08 Duke University Chimeric molecule and nucleic acid encoding same
US20070020722A1 (en) * 2003-04-30 2007-01-25 Counter Christopher M Telomere elongation
US20040248246A1 (en) * 2003-06-05 2004-12-09 The Government Of The United States Compositions and methods for enhancing differential expression
US7598077B2 (en) 2003-06-05 2009-10-06 The United States Of America As Represented By The Department Of Health And Human Services Compositions and methods for enhancing differential expression
US20060052324A1 (en) * 2004-08-05 2006-03-09 Artandi Steven E Methods and compositions for cell activation
US10383928B2 (en) 2010-02-16 2019-08-20 Ultimovacs As Telomerase polypeptide vaccine for treating cancer
US11529403B2 (en) 2010-02-16 2022-12-20 Ultimovacs As Telomerase polypeptide vaccine for treating cancer
US10251944B2 (en) 2016-01-19 2019-04-09 Pfizer Inc. Cancer vaccines
US11058753B2 (en) 2016-01-19 2021-07-13 Pfizer Inc. Cancer vaccines

Also Published As

Publication number Publication date
US20060275267A1 (en) 2006-12-07
US7091021B2 (en) 2006-08-15
US20020102686A1 (en) 2002-08-01
AU3375299A (en) 1999-10-18
WO1999050386A2 (en) 1999-10-07
US20150087064A1 (en) 2015-03-26
WO1999050386A3 (en) 2000-02-17
US8796438B2 (en) 2014-08-05

Similar Documents

Publication Publication Date Title
US6337200B1 (en) Human telomerase catalytic subunit variants
JP3869092B2 (en) Human telomerase catalytic subunit
EP2258383B1 (en) Antigenic peptides derived from telomerase
WO1998014592A9 (en) Telomerase reverse transcriptase
AU745420B2 (en) Human catalytic telomerase sub-unit and its diagnostic and therapeutic use
AU742489B2 (en) Regulatory DNA sequences of the human catalytic telomerase sub-unit gene, diagnostic and therapeutic use thereof
US20080050809A1 (en) Novel human kinases and polynucleotides encoding the same
Ogawa et al. Human liver serine dehydratase: cDNA cloning and sequence homology with hydroxyamino acid dehydratases from other sources
US6734009B2 (en) Human kinases and polynucleotides encoding the same
US6476210B2 (en) Human kinases and polynucleotides encoding the same
US6777221B2 (en) Human proteases and polynucleotides encoding the same
May et al. Methotrexate dose-escalation studies in transgenic mice and marrow transplant recipients expressing drug-resistant dihydrofolate reductase activity.
US6806073B2 (en) Human kinase proteins and polynucleotides encoding the same
EP0852619B1 (en) A modified glutamic acid decarboxylase (gad)
US6720173B1 (en) Human kinase protein and polynucleotides encoding the same
US20020147320A1 (en) Novel human kinase proteins and polynucleotides encoding the same
US6773906B2 (en) Human kinase and polynucleotides encoding the same
US6716616B1 (en) Human kinase proteins and polynucleotides encoding the same
US20030008365A1 (en) Novel human kinases and polynucleotides encoding the same
US6815188B2 (en) Human kinases and polynucleotides encoding the same
Molina Studies of mammalian uracil-DNA glycosylase and base excision repair
Yang Analyses of terminal deoxynucleotidyl transferase: Probing the enzyme active site and gene promoter

Legal Events

Date Code Title Description
AS Assignment

Owner name: GERON CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MORIN, GREGG B.;REEL/FRAME:009567/0511

Effective date: 19981027

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF COLORADO, COLORAD

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GERON CORPORATION;REEL/FRAME:017776/0812

Effective date: 20060508

Owner name: GERON CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GERON CORPORATION;REEL/FRAME:017776/0812

Effective date: 20060508

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12